REVIEW article

Front. Mol. Neurosci., 04 July 2025

Sec. Pain Mechanisms and Modulators

Volume 18 - 2025 | https://doi.org/10.3389/fnmol.2025.1625943

Exosome-mediated miRNA delivery: a molecular switch for reshaping neuropathic pain therapy

Ziqing Wei,&#x;&#x;Ziqing Wei1,2Chunhui Guo,&#x;&#x;Chunhui Guo1,2Hang Zhou,&#x;Hang Zhou1,2Yanling Wu,&#x;Yanling Wu1,2Xudong ZhouXudong Zhou3Jibing Chen
&#x;Jibing Chen2*Fujun Li,
&#x;Fujun Li2,4*
  • 1Graduate School, Guangxi University of Chinese Medicine, Nanning, China
  • 2Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
  • 3Graduate School, Guangxi University, Nanning, China
  • 4Zhuhai Maternal and Child Health Care Hospital, (Guangxi University of Chinese Medicine), Zhuhai, China

Neuropathic pain (NP) is a chronic condition caused by nerve injury or disease. It remains a therapeutic challenge because conventional drugs have limited efficacy and cause adverse effects. Exosomes, with the ability to cross the blood-brain barrier, low immunogenicity, and tissue-homing capacity, have emerged as promising nanovehicles for precise microRNA (miRNA) delivery to modulate key NP pathologies such as neuroinflammation, neuronal hyperexcitability, mechanical allodynia, and thermal hyperalgesia. In this review, we highlight recent advances in exosome-mediated miRNA therapy for NP. We also elucidate the molecular mechanisms and unique advantages of exosomes as both delivery platforms and intrinsic therapeutic agents. We synthesize evidence from preclinical models and initial clinical-stage studies, addressing translational challenges in scalable production and targeted delivery. Through sustained innovation and multidisciplinary collaboration, exosome-based miRNA delivery systems demonstrate transformative potential to overcome current therapeutic limitations, enabling novel NP management strategies.

1 Introduction

Neuropathic pain (NP) constitutes a debilitating chronic condition originating from lesions or diseases affecting the somatosensory nervous system (Finnerup et al., 2021). Afflicting approximately 7% of the global population, it imposes substantial clinical, economic, and societal burdens (Savelieff et al., 2025). Patients typically experience characteristic manifestations including spontaneous burning sensations, electric shock-like pain, and mechanically evoked allodynia—paradoxical pain perception in response to non-noxious stimuli such as light touch. The pathophysiology involves multifaceted mechanisms encompassing peripheral and central sensitization (e.g., dysregulated ion channels, NMDA receptor activation), neuroinflammation (e.g., TLR4/NF-κB signaling in glial cells), and impaired neural repair (Vranken, 2012; Zhang et al., 2023; Kaye et al., 2024).

Current NP management primarily relies on pharmacological interventions (e.g., antidepressants, calcium channel modulators, topical anesthetics) (Alcántara-Montero and Pacheco-de Vasconcelos, 2022; Bayer et al., 2004; Moisset et al., 2022) and neuromodulation techniques [e.g., transcutaneous electrical nerve stimulation (TENS), spinal cord stimulation (SCS)] (da Silva et al., 2024; Leung et al., 2020). However, these approaches face critical limitations: systemic adverse effects (sedation, anticholinergic effects), insufficient efficacy in subsets of patients, and inability to concurrently target multiple NP mechanisms. Compromised central nervous system bioavailability due to the blood-brain barrier (BBB) further restricts therapeutic effectiveness (Amescua-Garcia et al., 2018; Ardeleanu et al., 2020). Consequently, NP remains inadequately managed in many patients, underscoring the urgent need for novel multitargeted therapeutic strategies with enhanced central nervous system (CNS) delivery capabilities.

MicroRNAs (miRNAs), serving as pivotal post-transcriptional regulators in NP pathogenesis, modulate neuroinflammation, neuronal hyperexcitability, and nerve repair (Wang et al., 2024). Their capacity to concurrently fine-tune multiple target gene networks offers significant therapeutic advantages over single-target drugs (Chang et al., 2017; Peng et al., 2017). Nevertheless, clinical translation of miRNA therapies is hindered by rapid degradation, poor cellular uptake, and inefficient BBB penetration (Shityakov et al., 2022). Exosomes—natural nanoscale extracellular vesicles—provide a promising solution to these delivery challenges. They inherently protect cargo molecules (e.g., miRNAs) from degradation, exhibit low immunogenicity, possess intrinsic homing capacity toward injured tissues, and critically, traverse the BBB (Liu et al., 2024). Recent advances in exosome engineering further enhance their delivery potential (Nouri et al., 2024). Exosome-mediated miRNA delivery thus represents a transformative strategy to overcome limitations of conventional NP treatments through precision modulation of multiple pathological mechanisms. This review focuses on the emerging paradigm of exosome-mediated miRNA delivery for NP management. We outline the therapeutic rationale for miRNAs in NP pathogenesis and exosomes’ unique advantages as delivery vehicles, evaluate preclinical and clinical evidence for exosomal miRNA efficacy, and discuss clinical translation challenges and future directions.

2 Methods

This narrative review synthesizes literature retrieved from electronic databases including PubMed and ClinicalTrials.gov using core search terms: “exosomes,” “microRNA,” “neuropathic pain,” and “miRNA pain therapy” with their English equivalents. Included studies fulfilled these criteria: (1) mechanistic validation in animal or cellular models, and (2) documented evidence of exosome-mediated miRNA delivery. Exclusion criteria comprised case reports and non-English publications.

2.1 miRNAs in NP pathogenesis

2.1.1 Expression profiles and therapeutic potential

MicroRNAs (miRNAs) are endogenous small non-coding RNAs (approximately 22 nucleotides) that regulate gene expression by binding to the 3′ untranslated region (3′ UTR) of target mRNAs. This interaction, primarily mediated by a 6–8 nucleotide seed sequence, induces mRNA degradation or translational repression (Liu et al., 2025; Sun et al., 2017; Yang X. et al., 2023). Through this post-transcriptional regulation, miRNAs orchestrate critical cellular processes in NP pathogenesis, including neuroinflammation, neuronal hyperexcitability, and impaired nerve repair (Wilkerson et al., 2020). miRNA-mRNA network analysis identified multiple dysregulated miRNAs (e.g., miR-30c-5p, miR-16-5p) and their target genes (Rnase4, Egr2), revealing inflammation-associated regulatory mechanisms in NP (Cai et al., 2020). These findings support their potential as diagnostic biomarkers and therapeutic targets. Widespread miRNA alterations occur at key pain-processing sites [dorsal root ganglia (DRG) and spinal cord] across NP models [spared nerve injury (SNI), spinal nerve ligation (SNL), chronic constriction injury (CCI), and diabetic neuropathy]. Specific miRNAs including miR-21, miR-124, and miR-146a demonstrate significant dysregulation following nerve injury (Zhong et al., 2019; Zhang et al., 2019; Lv et al., 2017). Consistent with these observations, clinical studies report abnormal expression of miR-21, miR-146a, and miR-155 in sural nerves, skin biopsies, and circulating leukocytes from patients with painful peripheral neuropathy (Leinders et al., 2017). Mechanistically, these dysregulated miRNAs converge on three core pathways: neuroinflammatory signaling modulation (TLR4/NF-κB, NLRP3 inflammasome, cytokine release via TRAF6/IRAK1/STAT3 targeting), ion channel regulation (Nav1.7, TRPV1, Kv channels), and mediation of neuroimmune interactions and neural repair processes (Yan et al., 2017; Sun et al., 2021).

The extensive dysregulation of miRNAs establishes them as potent therapeutic targets for NP, offering three primary advantages:

1. Multi-target potential: Individual miRNAs regulate gene networks governing NP mechanisms (e.g., neuroinflammation, excitability), surpassing single-target drugs.

2. Mechanism-driven efficacy: Functional restoration of specific miRNAs (e.g., miR-146a-5p) using agomirs/antagomirs alleviates pain hypersensitivity and neuroinflammation in preclinical models (Wang et al., 2018).

3. Diagnostic and predictive biomarkers: Distinct miRNA expression patterns in biofluids or tissues (e.g., elevated serum hsa-miR-19a-3p and hsa-miR-19b-3p) enable NP subtype stratification, informing personalized therapies (Tavares-Ferreira et al., 2019).

2.1.2 Core regulatory mechanisms

2.1.2.1 Regulation of inflammatory cascades

Within NP pathology, miRNAs exert precise control over neuroimmune interactions by targeting critical signaling nodes: During inflammation initiation, Toll-like receptor (TLR) family members including TLR4 recognize damage-associated molecular patterns (DAMPs), activating the IRAK1/TRAF6 complex through MyD88-dependent pathways. This drives NF-κB/MAPK activation and progressive release of pro-inflammatory cytokines including TNF-α and IL-1β (Zarezadeh Mehrabadi et al., 2022). During inflammation amplification, miR-23a inhibits IL-1β maturation in microglia by dual-targeting CXCR4 (immune cell chemokine receptor) and TXNIP (key NLRP3 activation factor) (Pan et al., 2018). Concurrently, the CXCL12/CXCR4 axis recruits immune cells such as macrophages to infiltrate injury sites, exacerbating neuroinflammation (Liu et al., 2019), whereas miR-144 and miR-140 suppress this pathway (Li et al., 2021; Zhang et al., 2020a). Through negative feedback regulation, miR-146a-5p binds the 3′ UTR of TRAF6/IRAK1 mRNAs, establishing self-limiting control of NF-κB activation (Hou et al., 2021). Additionally, miRNAs modulate inflammatory cascades by targeting transcription factors: miR-136 and miR-128-3p inhibit ZEB1, blocking pro-inflammatory gene transcription (Bao et al., 2018; Shen et al., 2019; Yan et al., 2018; Zhang et al., 2020b). miR-363-5p targets SERPING1 (regulated by SP5), conferring dual analgesic/anti-inflammatory effects that SERPING1 overexpression negates (Wu et al., 2025). Collectively, miRNAs orchestrate multi-layered regulation of neuroinflammation, establishing them as potential therapeutic targets for NP.

2.1.2.2 Regulation of ion channel homeostasis

Neuronal hyperexcitability constitutes a core feature of NP, primarily driven by dysfunctional ion channel expression and activity. Exosome-mediated miRNA delivery enables novel therapeutic strategies for targeting channelopathies in voltage-gated sodium (Nav), potassium (Kv), calcium (Cav), and transient receptor potential (TRP) channels. Abnormal Nav isoform activation including Nav1.3, Nav1.7, and Nav1.8 can be selectively regulated through miRNAs: miR-30b directly targets SCN3A mRNA (Su et al., 2017); miR-182 inhibits SCN9A translation (Cai et al., 2018); and miR-7a downregulates β2 subunit (SCN2B) expression (Sakai et al., 2013), collectively reducing ectopic discharges in DRG. In contrast, miR-3584-5p exacerbates chronic constriction injury pain through Nav1.8 current suppression (Yang R. et al., 2023). Voltage-gated potassium channels critically control neuronal excitability by governing action potential generation, firing frequency, and neurotransmitter release (Manville et al., 2018; Kim and Nimigean, 2016). The miR-17-92 cluster maintains mechanical hypersensitivity post-injury through coordinated Kv regulation. This cluster contains six members—miR-17, miR-18a, miR-19a, miR-20a, miR-19b, and miR-92a-that remain persistently upregulated in injured sensory neurons (Sakai et al., 2017). Dysregulation mechanisms include miR-19a-mediated Kv4.2/4.3 mRNA targeting that reduces A-type potassium currents, and miR-137-induced Kcna2 inhibition that decreases Kv1.2 expression, both elevating neuronal excitability and pain perception. miR-137 inhibition restores Kv1.2 expression, normalizes neuronal excitability, and alleviates pain (Zhang et al., 2021). In calcium channel regulation, miR-103 targets CACNB1/CACNA2D1 (Cav1.2 auxiliary subunits) (Favereaux et al., 2011) while miR-32-5p silences Cav3.2 through histone methylation (Qi et al., 2022), both reducing calcium influx to block nociceptive sensitization. TRP channel modulation involves miR-375 and miR-455 suppressing TRPV1 expression (Li Z. et al., 2022), whereas miR-141-5p alleviates oxaliplatin-induced NP by inhibiting TRPA1 expression, thereby reducing Ca2+ influx and neuronal excitability (Zhang and Chen, 2021).

2.1.2.3 Neural regeneration and repair

Persistent NP following nerve injury may cause cellular damage or neuronal death in spinal cord and peripheral nerve tissues (Cohen et al., 2021). Recent studies reveal that injured peripheral neurons release endogenous neurotrophic factors including brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and nerve growth factor (NGF), promoting neuronal survival and axonal regeneration (Keefe et al., 2017; Han and Xu, 2020). The bidirectional regulatory capacity of exosomal miRNAs offers unique therapeutic value for neural repair. Excessive glial scar formation impedes axonal regeneration. miRNAs balance pro-inflammatory and repair processes by targeting glial activation states: miR-503-5p alleviates NP in type 2 diabetes mellitus (T2DM) mice through suppressing SEPT9 expression in astrocytes, while miR-204 enhances sensory functional recovery by upregulating glial cell-derived neurotrophic factor (GDNF) in microglia (Guo et al., 2024; Shen et al., 2020). Concurrently, miRNAs directly regulate neuronal regeneration: miR-155 deficiency promotes axonal regeneration through enhanced SPRR1A expression (Gaudet et al., 2016), whereas miR-135a and miR-135b counteract regenerative inhibition by suppressing Kruppel-like factor 4 (KLF4) (van Battum et al., 2018). miR-210 inhibits apoptosis via ephrin-A3 (EFNA3) to support neuronal survival (Hu et al., 2016). Notably, certain miRNAs exhibit dual regulatory properties. For example, miR-21 promotes Schwann cell-mediated axonal regeneration by inhibiting EPHA4/TIMP3 (Ning et al., 2020), yet activates the epidermal growth factor receptor (EGFR) pathway to exacerbate glial scarring (Kar et al., 2021). Across various neural injury models, miR-21 demonstrates functional versatility by modulating multiple signaling pathways to promote neural repair (Ning et al., 2020; Kar et al., 2021; Li et al., 2018).

These findings demonstrate that miRNAs therapeutically target core NP mechanisms (Table 1). However, clinical translation faces challenges including nuclease-mediated degradation and inadequate targeting specificity (Pedder et al., 2025; Wang and Wu, 2024). Developing stable delivery systems for miRNA mimics/inhibitors remains a critical unmet need (Zhang et al., 2017).

Table 1
www.frontiersin.org

Table 1. Core mechanisms of miRNA targeted intervention in neuropathic pain.

2.2 Overview of exosomes

2.2.1 Biological properties

Exosomes represent a subtype of extracellular vesicles (EVs) ranging from 50–150 nm in diameter. They are distinguishable from microvesicles (100–1,000 nm) by surface markers CD63/CD9 (Meldolesi, 2018). These vesicles form through bilayer invagination of cellular membranes and are subsequently released from multivesicular bodies (MVBs) (Yang et al., 2024). Though initially considered cellular waste disposal machinery, exosomes secreted by all cell types—including immune cells and neurons—are now recognized as crucial mediators of intercellular and intracellular communication. MVBs reside within cell bodies of DRG sensory neurons, which may release EVs (including exosomes) under appropriate conditions. Exosomes contain functionally diverse proteins essential for cell adhesion, membrane fusion, metabolism, and signal transduction. Beyond proteins, they carry multiple nucleic acids including miRNAs, messenger RNAs (mRNAs), DNA fragments, and long non-coding RNAs. These constituents mediate intercellular signaling in biological processes such as immune modulation and neural transmission (Bahram Sangani et al., 2021). Notably, exosomes are widely present in bodily fluids and transmit molecular signals via paracrine, autocrine, or endocrine pathways (Chen et al., 2019). Their biogenesis occurs in virtually all cell types (Habib et al., 2023), with particularly active production observed in tumor cells (Graner, 2019), immune cells, and neural cells. Emerging evidence indicates cellular origin critically determines both exosomal cargo composition and biological functionality (Yang et al., 2025). Substantial differences in contents, surface markers, and functions exist among exosomes derived from distinct cell types, suggesting specialized roles in biological processes.

2.2.2 Biogenesis and secretion processes

Though several mechanisms of exosome biosynthesis and secretion have been identified, many aspects remain incompletely understood. Exosome formation constitutes a complex multistep process involving membrane budding, invagination, multivesicular body (MVB) formation, and ultimate secretion (Ren et al., 2024). Recent studies demonstrate that exosome generation primarily relies on the intraluminal vesicle (ILV) formation pathway (Ghosh et al., 2024), comprising both ESCRT-dependent and ESCRT-independent mechanisms (Bavafa et al., 2025). The ESCRT (endosomal sorting complexes required for transport) complexes drive ILV generation through membrane remodeling and cargo sorting (Larios et al., 2020). Specifically, the ESCRT-0 complex recognizes and recruits cargo proteins, while ESCRT-I and ESCRT-II collectively promote membrane invagination, and ESCRT-III mediates vesicle fission and release (Ju et al., 2021). Precise regulation of the ESCRT machinery is critical not only for ILV formation but also for exosome secretion (Horbay et al., 2022). Beyond ESCRT-dependent pathways, exosome biogenesis involves alternative mechanisms (Yang et al., 2024). Lipid molecules including ceramide play pivotal roles by altering membrane lipid composition, enhancing fluidity, and facilitating ILV generation (Yanagawa et al., 2024). Furthermore, phosphatidylinositol 3-kinase (PI3K) and its product phosphatidylinositol-3,4,5-trisphosphate (PIP3) significantly contribute to exosome production (Yao et al., 2025). Exosome trafficking and release constitute equally complex processes governed by molecular regulators such as Rab GTPases (e.g., Rab27a/b) (Kim et al., 2024). These GTPases regulate MVB-plasma membrane fusion to ensure precise exosome transport and secretion. Recipient cells internalize exosomes primarily through endocytosis, direct membrane fusion, or surface receptor interactions (Vučemilović, 2024). Though endocytosis represents the predominant mechanism, direct fusion offers greater therapeutic promise for drug delivery due to enhanced intracellular cargo release efficiency (Hushmandi et al., 2024). These discoveries deepen our understanding of exosome biogenesis while establishing theoretical foundations for novel therapeutic strategies.

2.3 Advantages of exosomes as delivery vehicles

2.3.1 Natural targeting capacity

The targeting capacity of exosomes primarily depends on their surface characteristics and molecular cargo. Research confirms that multiple specific proteins on exosomal surfaces mediate interactions with target cells. Notably, lysosome-associated membrane glycoprotein 2B (LAMP2B) enhances exosomal binding to neurons and their subsequent internalization, establishing its role as a key targeting protein (Qiao et al., 2023). This property provides natural targeting advantages for exosomal drug delivery. TGF-β1-induced human umbilical cord smooth muscle cell (hUCSMC)-derived exosomes exhibit effective targeting toward microglia, suppressing microglial hyperplasia and alleviating NP. Mechanistic studies reveal that urothelial cancer-associated 1 (UCA1) directly interacts with miR-95-5p to release FOXO3a expression (Mou et al., 2023). These natural targeting mechanisms enhance therapeutic efficacy while reducing impacts on non-target cells and minimizing potential side effects. Additionally, exosomes support targeting through neuronal communication functions. By binding directly to neurons and modulating their physiological states, exosomes influence pain perception and processing (Frühbeis et al., 2013). Following peripheral axonal injury, DRG sensory neurons release exosomes enriched with upregulated miR-21-5p. These exosomes are readily phagocytosed by macrophages, promoting pro-inflammatory polarization and inflammatory factor release. Intrathecal administration of miR-21 inhibitors prevents macrophage infiltration and NP development (Simeoli et al., 2017). Thus, tissue-specific exosome delivery circumvents detrimental neuron-macrophage communication, offering novel therapeutic opportunities for NP.

2.3.2 Ability to cross the blood-brain barrier

BBB comprises tight junctions between endothelial cells in brain capillaries, primarily protecting the CNS from harmful substances (Lerussi et al., 2025). However, this barrier also restricts entry of many therapeutics, complicating neurological disorder treatment (Gong et al., 2025). Consequently, identifying carriers capable of crossing the BBB has become a research priority. Exosomes emerge as ideal candidates due to their natural biocompatibility and low immunogenicity. Exosomes can traverse the BBB bidirectionally between bloodstream and brain, though specific mechanisms for peripheral-to-brain migration remain incompletely elucidated (Banks et al., 2020). Exosomes primarily cross the BBB via transcytosis-transporting through intracellular compartments similarly to immune cells and pathogens-rather than paracellular routes through extracellular spaces. Post-crossing, two functional possibilities exist: complete traversal of the endothelial barrier for global brain effects (Khan et al., 2022), or sequestration within brain endothelial cells influencing these cells and triggering specific transport mechanisms (Saeedi et al., 2019; Console et al., 2019). This transmigration capability extends beyond neural stem cell (NSC)-derived exosomes. Other exosomes, including those from bone marrow mesenchymal stem cells (MSCs) and placental tissue, demonstrate similar BBB-crossing capacities. Clinically, exosomes’ penetrative ability positions them as novel neurological therapeutics. MSC-derived exosomes alleviate NP in chronic models by suppressing microglial activation and reducing neuroinflammation (Gao et al., 2023b). Moreover, exosomes show potential for delivering therapeutic molecules including miRNAs and proteins that modulate pain-processing and inflammatory pathways (Kang and Guo, 2022; Di Ianni et al., 2025).

2.3.3 Low immunogenicity and high stability

Exosomal biocompatibility enables prolonged systemic circulation without immune recognition or clearance. This property permits effective therapeutic molecule delivery, enhancing efficacy while minimizing side effects. The membrane structure protects encapsulated bioactive components, maintaining stability in vivo and in vitro (Tang et al., 2021). Studies confirm prolonged exosomal circulation effectively avoids clearance by the reticuloendothelial system (Patil et al., 2020). Compared to traditional drug delivery systems, exosomes better preserve therapeutic activity and achieve higher concentrations in target tissues. Notably, surface molecules including CD47, CD24, CD44, and CD31 function as anti-phagocytic signals, helping exosomes evade phagocytic clearance by macrophages. This enhances systemic stability and bioavailability (Parada et al., 2021). As naturally derived carriers, exosomes show minimal long-term accumulation in organs compared to viral vectors, resulting in negligible systemic toxicity (Yang et al., 2015). Recent research reveals exosomes provide protection during thermal stress by transferring thermotolerance signals that help cells maintain viability under extreme conditions (Logan et al., 2024).

Exosomes constitute highly efficient miRNA delivery vehicles due to their biocompatibility, low immunogenicity, and rapid membrane fusion capacity (Bian et al., 2025). To systematically compare advantages, Table 2 details exosome-based delivery versus conventional therapies across targeting specificity, blood-brain barrier penetration, and side effects.

Table 2
www.frontiersin.org

Table 2. Comparison of exosome-based delivery systems vs. conventional therapies.

2.4 Exosome-mediated miRNA therapy: evidence and mechanisms

2.4.1 Preclinical model evidence: validation of efficacy and mechanism

2.4.1.1 Exosome-delivered miRNA targeting neuroinflammation: mechanisms underlying analgesia

Exosomal miRNAs exhibit high stability and amplification potential due to their lipid bilayer structure, enabling traversal across blood-brain or blood-spinal cord barriers to mediate analgesia in chronic pain models (Huh et al., 2017; Ding et al., 2019). In CCI rat models, miR-181c-5p expression is significantly downregulated, while intrathecal delivery of exosomal miR-181c-5p alleviates NP and neuroinflammatory responses (Zhang et al., 2022). Exosomal miRNAs operate through autocrine secretion and transport to target sites, acting on macrophages, microglia, neurons, or other tissue cells to regulate inflammatory factor secretion and oxidative stress, thereby modulating NP pathogenesis. In diabetic nephropathy (DN) mouse models, macrophage-derived EVs enriched with miR-21-5p enhance pyroptosis by upregulating A20 (a negative regulator of the NF-κB pathway). Correspondingly, intrathecal administration of anti-miR-21-5p antibodies reduces dorsal root ganglion DRG hyperalgesia and macrophage recruitment (Ding et al., 2021). Similarly, human umbilical cord mesenchymal stem cell (huc-MSC)-derived exosomes regulate microglial pyroptosis and autophagy through the miR-146a-5p/TRAF6 axis (Hua et al., 2022). Certain exosomal miRNAs alleviate neuroinflammation by inhibiting pro-inflammatory cytokine production or promoting anti-inflammatory cytokine release. For instance, human umbilical cord MSC-derived exosomes upregulate autophagy proteins LC3-II and beclin1 while blocking NLRP3 inflammasome activation via miR-146a-5p/TRAF6 signaling in the spinal dorsal horn (Hua et al., 2022). Bone marrow mesenchymal stem cell-derived extracellular vesicles (BMSC-EVs) enriched with miR-23b regulate TLR4/NF-κB signaling, attenuating inflammation and improving pathological status in SCI rats (Nie and Jiang, 2021). Additionally, BMSC-derived exosomes promote miR-145-5p expression to inhibit TLR4/NF-κB pathway activation, demonstrating significant anti-inflammatory and pathway regulatory effects in both SCI rats and PC12 cells (Jiang and Zhang, 2021). These findings highlight the therapeutic value of exosomal miRNAs in controlling neuroinflammatory signaling and ameliorating neural damage.

2.4.1.2 Synergistic protective mechanism of exosome-mediated miRNA in nerve regeneration and anti-apoptosis

Exosomes derived from Schwann cells, macrophages, and mesenchymal stem cells (MSCs) promote peripheral nerve regeneration (Sanchez et al., 2017; Marofi et al., 2017; Mead and Tomarev, 2017). Studies confirm exosomes facilitate regeneration of damaged nerves and improve motor function recovery in regenerated nerves in rat sciatic nerve compression models (Bucan et al., 2019). Exosomes enriched with the miR-17-92 cluster activate the PI3K/Akt/mTOR/GSK-3β signaling pathway by targeting PTEN, increasing neural plasticity and functional recovery (Xin et al., 2017). Specifically, skin precursor-derived Schwann cell extracellular vesicles (SKP-SC-EVs) containing miR-21-5p enhance DRG sensory neuron growth and survival through the PTEN-PI3K pathway (Cong et al., 2021). Simultaneously, miR-23b-3p promotes axonal regeneration by directly targeting Nrf1 mRNA (Xia et al., 2020). Regarding SCI models, intrathecal injection of MSC-derived exosomes significantly upregulates miR-99b-3p expression while activating microglial autophagy and alleviating mechanically induced allodynia caused by microglial activation (Gao et al., 2023a). Concurrently, exosomes from miR-126-modified MSCs reduce neuronal apoptosis while promoting functional regeneration (Huang et al., 2020), and exosomes derived from both MSCs and human neuroepithelial stem cells—enriched with miR-29b—downregulate PTEN and caspase-3 to inhibit neuronal apoptosis and confer therapeutic efficacy for SCI (Yu et al., 2019). Exosomal miR-499a-5p plays a neuroprotective role in SCI by targeting the JNK3/c-jun apoptotic pathway, reducing nerve cell apoptosis post-injury while decreasing cavity formation in lesioned areas. This mechanism promotes functional hindlimb recovery in rats through adipose-derived mesenchymal stem cell exosomes (ADSC-EXs) carrying miR-499a-5p, which reduce JNK3 expression and diminish nerve cell death after SCI (Liang et al., 2022). Adipose-derived mesenchymal stem cells (ADSCs) contain abundant neurotrophic factors, immunomodulatory factors, and angiogenic factors associated with neuronal differentiation and nerve regeneration (Harrell et al., 2022). Through miRNA transport, exosomes enhance neuronal cell activity and reduce apoptosis during early stages, thereby promoting functional recovery (see Figure 1).

Figure 1
Diagram illustrating a study on exosomal miRNA treatment in neuropathic pain models. Sources of miRNA include nerve, MSC, and immune cells. A mouse model receives an injection, showing reduced neuroinflammation, enhanced M2 polarization, decreased pain, promoted axon regeneration, inhibited neuron apoptosis, and recovered motor function.

Figure 1. Exosomal miRNA treatment for neuropathic pain. Schematic illustration of exosomal miRNA-based therapeutic strategies for neuropathic pain. Neuropathic pain models (e.g., CCI, SCI, DN) are subjected to exosomal miRNAs sourced from multiple cellular origins: nerve-resident cells (Schwann cells, microglia, neurons), mesenchymal stem cells (huc-MSCs, BMSCs, ADSCs), and immune/other lineages (macrophages, T cells, skin precursor cells). These exosomal miRNAs modulate neuropathic pain pathophysiology through dual-pronged mechanisms: ① anti-inflammatory actions (reducing neuroinflammation, promoting M2 polarization, decreasing pain sensitization); ② neuroregenerative effects (enhancing axon regeneration, suppressing neuron apoptosis, restoring motor function).

Thus, exosome-mediated miRNA transfer represents an effective therapeutic approach for NP (Table 3).

Table 3
www.frontiersin.org

Table 3. Exosomal miRNAs in preclinical models of neuropathic pain: therapeutic efficacy and mechanisms.

2.4.2 Clinical investigations

We identified clinical trials evaluating exosomes as therapeutic agents for NP (Table 4), including one published clinical trial and one ongoing registered trial (data current through June 2025). These investigations provide foundational insights into the complex mechanistic actions of exosomal miRNAs.

Table 4
www.frontiersin.org

Table 4. Clinical trials of exosome-based therapies for neuropathic pain.

The completed Phase I trial IRCT20200502047277N1 (Akhlaghpasand et al., 2024) adopted a single-center design focused on safety assessment rather than efficacy evaluation. Although preliminary improvements demonstrate clinical relevance, natural disease progression complicates definitive efficacy determination. Observed sensory-motor functional enhancements in certain patients may derive from either exosomal therapeutic effects or spontaneous disease resolution. This reflects methodological constraints in current efficacy evaluation approaches, necessitating larger multicenter randomized Phase II/III trials to validate outcomes and establish the definitive therapeutic role of exosome therapy in NP. Safety monitoring revealed no severe adverse events. However, long-term biological consequences of exosomes in vivo remain undetermined. Potential concerns include sustained biological activity, immune response induction, and interference with normal cellular functions. Notably, engineered exosomal products warrant particularly rigorous risk assessment due to greater uncertainty.

Regrettably, current clinical data demonstrate: limited study accessibility, data incompleteness (e.g., pending NCT05152368 results), and recurrent methodological limitations: small cohorts, abbreviated follow-up periods, and non-blinded designs lacking placebo controls. These constraints impede comprehensive scientific assessment.

2.5 Evolving paradigms in exosome delivery: administration routes and biomaterial innovations

2.5.1 Comparative analysis of exosome delivery routes

Delivery routes critically influence therapeutic efficacy due to their significant impacts on exosome distribution, absorption, and functional outcomes. Different administration methods—including intrathecal injection and intranasal instillation—distinctly affect exosome biodynamics. Intrathecal injection achieves targeted delivery to injury sites, maximizing local effects while minimizing systemic side effects. Epidural injection specifically targets spinal cord tissue with reduced complication rates. Intravenous administration remains the predominant preclinical delivery route (Hassanzadeh et al., 2021), offering systemic distribution with technical simplicity and lower complication risks via caudal vein injection.

Two registered clinical studies utilize intrathecal injection and intranasal instillation. Intranasal administration leverages olfactory and trigeminal axonal pathways to bypass the blood–brain barrier, enabling direct therapeutic delivery to brain tissue. Compared to invasive approaches (intrathecal or parenchymal delivery) with infection risks, this non-invasive technique offers significant advantages. Multiple preclinical studies confirm intranasally administered exosomes effectively prevent neuronal apoptosis and improve neurological recovery (Gotoh et al., 2025).

2.5.2 Engineering strategies for enhanced targeting

Exosomes exhibit unique advantages as natural drug carriers, but their clinical translation is limited by insufficient targeting specificity. For example, intravenous administration leads to rapid clearance by phagocytic organs such as the liver and spleen, significantly reducing target organ enrichment efficiency. This not only diminishes therapeutic efficacy but also raises risks of off-target toxicity. Novel exosome engineering techniques enhance delivery precision through customized miRNA loading and surface modifications (Del Pozo-Acebo et al., 2021). Research has developed genetically engineered exosomes carrying miR-21 combined with collagen-I (Col-I) scaffolds to repair SCI, demonstrating improved stability, delivery efficiency, and targeting (Liu et al., 2022).

Additionally, researchers encapsulated adipose tissue-derived mesenchymal stem cell exosomes (AD-MSC-EXs) within collagen and fibrin hydrogels (Afsartala et al., 2023), extending active retention at injury sites in SCI rat models. Gelatin sponge (Gelfoam)-loaded human umbilical cord mesenchymal stem cell exosomes (HucMSC-EXs) achieve precise delivery to SCI sites while promoting neural regeneration (Poongodi et al., 2024). Innovative tetrahedral DNA nanostructure (TDN)-based delivery systems incorporate RNase H-sensitive DNA–RNA hybrid sequences as bioswitches. Upon reaching target cells (e.g., in inflammatory or tumor microenvironments), RNase H specifically cleaves hybrid strands to trigger precise miRNA release (Li et al., 2025).

These engineering strategies can overcome biological barriers including phagocytic clearance and short half-life through surface functionalization, hydrogel sustained-release systems, and responsive nanoswitch designs, significantly enhancing spatiotemporal delivery precision.

2.6 Clinical translation: potential and challenges

2.6.1 Key challenges

How to address miRNA target gene complexity and functional validation bottlenecks?

How to predict exosomal miRNA therapeutic efficacy in individual patients?

What defines long-term in vivo distribution and safety profiles of therapeutic exosomes?

How to establish GMP-compliant large-scale exosome production?

How to optimize storage conditions to improve clinical feasibility?

How to reduce exosomal immunogenicity to enhance biological safety?

2.6.2 Complexity of miRNA target genes and challenges in functional validation

Neuropathic pain (NP) pathogenesis involves diverse cellular and molecular mechanisms. Different NP subtypes-including chemotherapy-induced, diabetic, and traumatic NP-exhibit distinct miRNA expression profiles and functional pathways. NP models demonstrate 2,776 differentially expressed RNA molecules comprising 219 miRNAs and 2,557 mRNAs. Crucially, miRNAs regulate multiple target genes simultaneously, frequently through indirect mechanisms, significantly complicating target identification (Li et al., 2019; Golmakani et al., 2024). Current bioinformatics tools predict potential targets but lack experimental validation, undermining prediction reliability. These regulatory interactions are further complicated by cell-type specificity, microenvironmental influences, and competitive binding with non-coding RNAs (e.g., lncRNAs and circRNAs) (Mukherjee et al., 2025). miRNA regulatory networks exhibit dynamic complexity, with target specificity varying across physiological and pathological states. For instance, while miR-133a-3p overexpression attenuates microglial activation and neuroinflammation in CCI models (Jia et al., 2021; Gao et al., 2024), its upregulation conversely promotes neuroinflammation and pain development in diabetic NP (DNP) models through TRAF6 and PIAS3 protein modulation (Chang et al., 2020). Accurately interpreting miRNA functions in neuropathology requires integrated analysis of both multi-target characteristics and cell-contextual functionality. Emerging technologies-particularly single-cell RNA sequencing, NGS, and machine learning algorithms (Picchio et al., 2025)-will likely advance miRNA-target identification, accelerating NP therapeutic development.

2.6.3 Scale-up: preparation, purification and quality control challenges

Exosome isolation employs diverse methods including ultracentrifugation, ultrafiltration, polymer precipitation, and immunoaffinity techniques. However, while ultracentrifugation remains the most common isolation technology, it demands specialized equipment and technical expertise while often causing damage to exosomes and functional loss (Baruah et al., 2024). Furthermore, exosome isolation is frequently contaminated by coexisting extracellular vesicles (such as microvesicles and apoptotic bodies), complicating purification processes and compromising analytical accuracy (Marjani et al., 2024). Exosomes contain diverse components including proteins, lipids, and RNAs, with biological activity closely linked to compositional integrity. Therefore, ensuring quality during preparation—particularly evaluating purity and bioactivity—represents an urgent challenge (Zhang F. et al., 2024). Currently, no standardized criteria exist to assess exosomal quality, hindering clinical translation (Sen et al., 2023; Fan et al., 2024). The International Society for Extracellular Vesicles (ISEV) aims to address these issues through its 2023 guidelines, providing technical guidance for documenting specific functional activities and procedural steps (Théry et al., 2018). Researchers are developing new technologies and standards: Microfluidics-based approaches enable improved isolation efficiency with reduced exosomal damage (Xing et al., 2025), while combined high-throughput analytical techniques and biological functional testing allow more comprehensive evaluation of exosomal quality and bioactivity (Zhang J. et al., 2024; Ishii and Tateno, 2025). These measures will enhance production consistency while ensuring clinical safety and efficacy. Future research should focus on developing efficient isolation technologies and rigorous quality control systems to achieve clinical translation of exosome-based therapies.

2.6.4 Stability and biosafety of delivery systems

Delivery system stability critically determines therapeutic duration and effects in vivo. Multiple studies demonstrate that exosomes exhibit optimal stability when stored at −80°C (Levy et al., 2023), though this condition proves impractical for routine clinical application. Optimizing storage conditions is essential to ensure reliability and effectiveness in clinical settings. Current research lacks sufficient data regarding the shelf life and in vivo stability of exosomal preparations (Palakurthi et al., 2024), limiting their clinical translation. Substantial technical challenges remain unresolved, including controversial issues surrounding administration routes, injection rates, and dosage standardization (Wang et al., 2023; Batrakova and Kim, 2015). Notably, in vivo studies show no significant positive correlation between exosome dosage levels and neuroregenerative outcomes, with higher doses failing to enhance therapeutic efficacy (Zhao et al., 2020). Certain delivery systems may activate immune responses, inducing inflammation or other adverse effects that compromise treatment effectiveness (Brain et al., 2021). Some researchers have utilized cell-targeted delivery systems (CDSEMs) constructed from edible materials (Li X. et al., 2022) and delivery systems fabricated using polymer matrices and other materials, which demonstrate outstanding performance in maintaining miRNA bioactivity and reducing immune responses in vivo, significantly enhancing biosafety (Poongodi et al., 2024). The biosafety of exosomes is significantly influenced by their cellular origin and purification processes. While exosomes derived from healthy cells demonstrate favorable safety profiles in vivo, those originating from pathological conditions may cause adverse reactions (Sohrabi et al., 2022; Yamayoshi et al., 2020). Comprehensive assessment of in vivo immune responses and potential side effects is mandatory before clinical application. Strict compliance with GMP (good manufacturing practice) standards during manufacturing processes is essential to mitigate patient risks. Metabolomics can identify off-target effects and adverse drug events by detecting early signs of drug-induced liver injury, cardiotoxicity, and other complications through metabolic profile analysis (Shaman, 2024; Røikjer et al., 2024). The incomplete functional characterization of exosomes hinders accurate prediction of their long-term safety and efficacy. Although surface modification with targeting peptides significantly enhances exosomal targeting capability, potential immunogenicity of these peptides raises concerns about immune responses in humans. Developing secure and effective methods for anchoring targeting peptides to exosomes thus remains a challenging pursuit.

3 Future directions

1. Integrating multi-omics technologies—including transcriptomics, proteomics, and metabolomics—will enable systematic elucidation exosomal miRNA mechanisms in NP. Researchers integrating genome-wide association studies (GWAS) with multi-omics data have revealed significant overlap in gene co-expression modules between NP and inflammatory pain (IP). Furthermore, integrated multi-omics analyses have identified specific miRNAs critically regulating neuroinflammation and neuronal excitability, while uncovering novel miRNA targets and signaling pathways (Ye et al., 2022). Leveraging omics technologies to select optimal donors and optimize exosome composition may consequently improve therapeutic outcomes (Lotfy et al., 2023).

2. Engineering exosomes through surface modifications (aptamers, antibodies, peptides) can enhance targeting precision to injured spinal cord regions and specific cell types, improving both delivery accuracy and therapeutic efficacy (Ye et al., 2023). Integrating exosomes with nanomedicine, materials science, and bioengineering could augment their therapeutic potential as delivery vehicles (Haroon et al., 2024). For instance, the RNAi-Tim3-Exo@SF hydrogel system delivers siRNA-Tim3-modified exosomes to precisely regulate Tim3 expression. This system stabilizes microtubules, promotes axonal regeneration, stimulates angiogenesis, modulates inflammatory microenvironments, and significantly improves motor function in spinal cord injury models. The key reparative mechanisms likely involve miR-155-5p within RNAi-Tim3-Exo (Dong et al., 2025). Such integrated strategies combining immunomodulation with tissue engineering may represent effective approaches for future clinical applications.

3. Current NP research minimally addresses large-animal models (pigs, non-human primates) in the literature. These species demonstrate greater neurological similarity to humans in axonal diameter, myelination patterns, and glial responses, enabling superior modeling of human pathological changes and pain behavior following neural injury (Karri et al., 2022). Consequently, the field urgently requires transitioning from exclusive rodent models to incorporating large-animal paradigms (e.g., porcine sciatic nerve injury models, non-human primate spinal nerve root compression models) (Ding et al., 2017). Such models better replicate human neuroanatomy and pain responses while enhancing preclinical pharmacodynamic predictability, providing robust platforms for developing targeted therapies.

4. Future exosome research should prioritize engineering exosomes specifically for drug delivery and clinical efficacy validation. Large-scale, multi-center studies with sufficient sample diversity and extended follow-up durations are essential to substantiate therapeutic efficacy and biosafety profiles (Figure 2).

Figure 2
Diagram depicting challenges and solutions in research and production segmented into four quadrants: 1. Basic Research: Highlights multi-omics integration, AI optimization, and single-cell sequencing versus gene complexity and validation tool insufficiency.2. Scalable Production: Features technology-driven isolation and regulatory synergy against inefficiencies and lack of standards.3. Delivery Systems: Discusses engineered targeting and immune evasion against low targeting efficiency and stability issues.4. Clinical Translation: Covers large animal models and regulatory collaboration versus animal model limitations and ethical gaps.Arrows indicate the flow between challenges and innovations.

Figure 2. Exosome-mediated miRNA delivery for neuropathic pain: design challenges and clinical pathways. Schematic of exosome design for neuropathic pain therapy, with challenges as the core. It maps hurdles across target gene complexity, production inefficiencies, delivery barriers, and clinical translation gaps to solutions (multi-omics, engineered targeting, regulatory pathways), illustrating how overcoming these accelerates exosome-mediated miRNA delivery to reshape NP treatment outcomes.

4 Conclusion

Neuropathic pain (NP) is a refractory disorder involving multiple pathological mechanisms. It presents new therapeutic opportunities through the regulatory efficacy of miRNAs. Exosomes serve as ideal miRNA carriers due to their endogenous stability and targeted delivery advantages. Preclinical evidence confirms exosome-mediated miRNA delivery effectively alleviates NP by modulating core signaling pathways. However, clinical translation faces persistent challenges including exosomal heterogeneity, delivery efficiency bottlenecks, and complexity of personalized treatments. Addressing these requires multidisciplinary convergence of exosome engineering, biomaterials science, and clinical validation to accelerate reliable therapeutic solutions. Although current clinical implementation remains nascent, ongoing research strongly supports their translational potential.

Author contributions

ZW: Writing – original draft, Writing – review & editing. CG: Writing – original draft, Writing – review & editing. HZ: Writing – original draft. YW: Writing – review & editing, Supervision. XZ: Validation, Supervision, Writing – original draft. JC: Funding acquisition, Resources, Project administration, Writing – review & editing. FL: Resources, Writing – review & editing, Funding acquisition, Project administration.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. This work was supported by the Regional Fund Project of the National Natural Science Foundation of China (No. 82460874), the General Program of Guangxi Natural Science Foundation (2024GXNSFAA010354), the Key Research and Development Program of Guangxi (Task No. Guike AB25069271), and the General Program of Guangxi Natural Science Foundation (Task No. 2025GXNSFAA0691007).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The authors declare that no Gen AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Afsartala, Z., Hadjighassem, M., Shirian, S., Ebrahimi-Barough, S., Gholami, L., Parsamanesh, G., et al. (2023). The effect of collagen and fibrin hydrogels encapsulated with adipose tissue mesenchymal stem cell-derived exosomes for treatment of spinal cord injury in a rat model. Iran. J. Biotechnol. 21:e3505. doi: 10.30498/ijb.2023.362229.3505

PubMed Abstract | Crossref Full Text | Google Scholar

Akhlaghpasand, M., Tavanaei, R., Hosseinpoor, M., Yazdani, K. O., Soleimani, A., Zoshk, M. Y., et al. (2024). Safety and potential effects of intrathecal injection of allogeneic human umbilical cord mesenchymal stem cell-derived exosomes in complete subacute spinal cord injury: a first-in-human, single-arm, open-label, phase I clinical trial. Stem Cell Res Ther 15:264. doi: 10.1186/s13287-024-03868-0

PubMed Abstract | Crossref Full Text | Google Scholar

Alcántara-Montero, A., and Pacheco-de Vasconcelos, S. R. (2022). Pharmacological approach to neuropathic pain: past, present and future. Rev. Neurol. 74, 269–279. doi: 10.33588/rn.7408.2021381

PubMed Abstract | Crossref Full Text | Google Scholar

Amescua-Garcia, C., Colimon, F., Guerrero, C., Jreige Iskandar, A., Berenguel Cook, M., Bonilla, P., et al. (2018). Most relevant neuropathic pain treatment and chronic low back pain management guidelines: a change pain Latin America advisory panel consensus. Pain Med. 19, 460–470. doi: 10.1093/pm/pnx198

PubMed Abstract | Crossref Full Text | Google Scholar

Ardeleanu, V., Toma, A., Pafili, K., Papanas, N., Motofei, I., Diaconu, C. C., et al. (2020). Current pharmacological treatment of painful diabetic neuropathy: a narrative review. Medicina 56:25. doi: 10.3390/medicina56010025

PubMed Abstract | Crossref Full Text | Google Scholar

Arthur, J. A., Reddy, A., Popat, U., Halm, J., Vaughan-Adams, N., Myers, A., et al. (2025). Abuse potential and analgesic efficacy of intravenous hydromorphone bolus administration among hospitalized patients with cancer pain: a double-blind, double dummy, randomized crossover trial. Cancer 131:e35723. doi: 10.1002/cncr.35723

PubMed Abstract | Crossref Full Text | Google Scholar

Bahram Sangani, N., Gomes, A. R., Curfs, L. M. G., and Reutelingsperger, C. P. (2021). The role of extracellular vesicles during CNS development. Prog. Neurobiol. 205:102124. doi: 10.1016/j.pneurobio.2021.102124

PubMed Abstract | Crossref Full Text | Google Scholar

Banks, W. A., Sharma, P., Bullock, K. M., Hansen, K. M., Ludwig, N., and Whiteside, T. L. (2020). Transport of extracellular vesicles across the blood-brain barrier: brain pharmacokinetics and effects of inflammation. Int. J. Mol. Sci. 21:4407. doi: 10.3390/ijms21124407

PubMed Abstract | Crossref Full Text | Google Scholar

Bao, Y., Wang, S., Xie, Y., Jin, K., Bai, Y., and Shan, S. (2018). MiR-28-5p relieves neuropathic pain by targeting Zeb1 in CCI rat models. J. Cell. Biochem. 119, 8555–8563. doi: 10.1002/jcb.27096

PubMed Abstract | Crossref Full Text | Google Scholar

Baruah, H., Sarma, A., Basak, D., and Das, M. (2024). Exosome: From biology to drug delivery. Drug Deliv. Transl. Res. 14, 1480–1516. doi: 10.1007/s13346-024-01515-y

PubMed Abstract | Crossref Full Text | Google Scholar

Batrakova, E. V., and Kim, M. S. (2015). Using exosomes, naturally-equipped nanocarriers, for drug delivery. J. Control. Release 219, 396–405. doi: 10.1016/j.jconrel.2015.07.030

PubMed Abstract | Crossref Full Text | Google Scholar

Bavafa, A., Izadpanahi, M., Hosseini, E., Hajinejad, M., Abedi, M., Forouzanfar, F., et al. (2025). Exosome: an overview on enhanced biogenesis by small molecules. Naunyn Schmiedeberg’s Arch. Pharmacol. 398, 6473–6508. doi: 10.1007/s00210-024-03762-9

PubMed Abstract | Crossref Full Text | Google Scholar

Bayer, K., Ahmadi, S., and Zeilhofer, H. U. (2004). Gabapentin may inhibit synaptic transmission in the mouse spinal cord dorsal horn through a preferential block of P/Q-type Ca2+ channels. Neuropharmacology 46, 743–749. doi: 10.1016/j.neuropharm.2003.11.010

PubMed Abstract | Crossref Full Text | Google Scholar

Bian, X., Zhou, L., Luo, Z., Liu, G., Hang, Z., Li, H., et al. (2025). Emerging delivery systems for enabling precision nucleic acid therapeutics. ACS Nano 19, 4039–4083. doi: 10.1021/acsnano.4c11858

PubMed Abstract | Crossref Full Text | Google Scholar

Brain, D., Plant-Hately, A., Heaton, B., Arshad, U., David, C., Hedrich, C., et al. (2021). Drug delivery systems as immunomodulators for therapy of infectious disease: relevance to COVID-19. Adv. Drug Deliv. Rev. 178:113848. doi: 10.1016/j.addr.2021.113848

PubMed Abstract | Crossref Full Text | Google Scholar

Bucan, V., Vaslaitis, D., Peck, C. T., Strauß, S., Vogt, P. M., and Radtke, C. (2019). Effect of exosomes from rat adipose-derived mesenchymal stem cells on neurite outgrowth and sciatic nerve regeneration after crush injury. Mol. Neurobiol. 56, 1812–1824. doi: 10.1007/s12035-018-1172-z

PubMed Abstract | Crossref Full Text | Google Scholar

Cai, W., Zhao, Q., Shao, J., Zhang, J., Li, L., Ren, X., et al. (2018). MicroRNA-182 alleviates neuropathic pain by regulating Nav1.7 following spared nerve injury in rats. Sci. Rep. 8:16750. doi: 10.1038/s41598-018-34755-3

PubMed Abstract | Crossref Full Text | Google Scholar

Cai, G., Zhu, Y., Zhao, Y., Chen, J., Guo, C., Wu, F., et al. (2020). Network analysis of miRNA and mRNA changes in the prelimbic cortex of rats with chronic neuropathic pain: pointing to inflammation. Front. Genet. 11:612. doi: 10.3389/fgene.2020.00612

PubMed Abstract | Crossref Full Text | Google Scholar

Chang, H. L., Wang, H. C., Chunag, Y. T., Chou, C. W., Lin, I. L., Lai, C. S., et al. (2017). miRNA expression change in dorsal root ganglia after peripheral nerve injury. J. Mol. Neurosci. 61, 169–177. doi: 10.1007/s12031-016-0876-7

PubMed Abstract | Crossref Full Text | Google Scholar

Chang, L. L., Wang, H. C., Tseng, K. Y., Su, M. P., Wang, J. Y., Chuang, Y. T., et al. (2020). Upregulation of miR-133a-3p in the sciatic nerve contributes to neuropathic pain development. Mol. Neurobiol. 57, 3931–3942. doi: 10.1007/s12035-020-01999-y

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, B. Y., Sung, C. W., Chen, C., Cheng, C. M., Lin, D. P., Huang, C. T., et al. (2019). Advances in exosomes technology. Clin. Chim. Acta 493, 14–19. doi: 10.1016/j.cca.2019.02.021

PubMed Abstract | Crossref Full Text | Google Scholar

Cohen, S. P., Vase, L., and Hooten, W. M. (2021). Chronic pain: an update on burden, best practices, and new advances. Lancet 397, 2082–2097. doi: 10.1016/S0140-6736(21)00393-7

PubMed Abstract | Crossref Full Text | Google Scholar

Cong, M., Shen, M., Wu, X., Li, Y., Wang, L., He, Q., et al. (2021). Improvement of sensory neuron growth and survival via negatively regulating PTEN by miR-21-5p-contained small extracellular vesicles from skin precursor-derived Schwann cells. Stem Cell Res Ther 12:80. doi: 10.1186/s13287-020-02125-4

PubMed Abstract | Crossref Full Text | Google Scholar

Console, L., Scalise, M., and Indiveri, C. (2019). Exosomes in inflammation and role as biomarkers. Clin. Chim. Acta 488, 165–171. doi: 10.1016/j.cca.2018.11.009

PubMed Abstract | Crossref Full Text | Google Scholar

da Silva, M. D. V., Martelossi-Cebinelli, G., Yaekashi, K. M., Carvalho, T. T., Borghi, S. M., Casagrande, R., et al. (2024). A narrative review of the dorsal root ganglia and spinal cord mechanisms of action of neuromodulation therapies in neuropathic pain. Brain Sci. 14:589. doi: 10.3390/brainsci14060589

PubMed Abstract | Crossref Full Text | Google Scholar

Del Pozo-Acebo, L., Hazas, M. L. L., Tomé-Carneiro, J., Gil-Cabrerizo, P., San-Cristobal, R., Busto, R., et al. (2021). Bovine milk-derived exosomes as a drug delivery vehicle for miRNA-based therapy. Int. J. Mol. Sci. 22:1105. doi: 10.3390/ijms22031105

PubMed Abstract | Crossref Full Text | Google Scholar

Di Ianni, E., Obuchi, W., Breyne, K., and Breakefield, X. O. (2025). Extracellular vesicles for the delivery of gene therapy. Nat. Rev. Bioeng. 3, 360–373. doi: 10.1038/s44222-025-00277-7

Crossref Full Text | Google Scholar

Ding, S. Q., Chen, J., Wang, S. N., Duan, F. X., Chen, Y. Q., Shi, Y. J., et al. (2019). Identification of serum exosomal microRNAs in acute spinal cord injured rats. Exp. Biol. Med. 244, 1149–1161. doi: 10.1177/1535370219872759

PubMed Abstract | Crossref Full Text | Google Scholar

Ding, X., Jing, N., Shen, A., Guo, F., Song, Y., Pan, M., et al. (2021). MiR-21-5p in macrophage-derived extracellular vesicles affects podocyte pyroptosis in diabetic nephropathy by regulating A20. J. Endocrinol. Investig. 44, 1175–1184. doi: 10.1007/s40618-020-01401-7

PubMed Abstract | Crossref Full Text | Google Scholar

Ding, W., You, Z., Shen, S., Yang, J., Lim, G., Doheny, J. T., et al. (2017). An improved rodent model of trigeminal neuropathic pain by unilateral chronic constriction injury of distal infraorbital nerve. J. Pain 18, 899–907. doi: 10.1016/j.jpain.2017.02.427

PubMed Abstract | Crossref Full Text | Google Scholar

Dong, X., Lu, Y., Hu, Q., Zeng, C., Zheng, J., Huang, J., et al. (2025). Engineered exosome-loaded silk fibroin composite hydrogels promote tissue repair in spinal cord injury via immune checkpoint blockade. Small :e2412170. doi: 10.1002/smll.202412170

Crossref Full Text | Google Scholar

Fan, X., Zhang, Y., Liu, W., Shao, M., Gong, Y., Wang, T., et al. (2024). A comprehensive review of engineered exosomes from the preparation strategy to therapeutic applications. Biomater. Sci. 12, 3500–3521. doi: 10.1039/D4BM00558A

PubMed Abstract | Crossref Full Text | Google Scholar

Favereaux, A., Thoumine, O., Bouali-Benazzouz, R., Roques, V., Papon, M. A., Salam, S. A., et al. (2011). Bidirectional integrative regulation of Cav1.2 calcium channel by microRNA miR-103: role in pain. EMBO J. 30, 3830–3841. doi: 10.1038/emboj.2011.249

PubMed Abstract | Crossref Full Text | Google Scholar

Finnerup, N. B., Kuner, R., and Jensen, T. S. (2021). Neuropathic pain: from mechanisms to treatment. Physiol. Rev. 101, 259–301. doi: 10.1152/physrev.00045.2019

PubMed Abstract | Crossref Full Text | Google Scholar

Frühbeis, C., Fröhlich, D., Kuo, W. P., Amphornrat, J., Thilemann, S., Saab, A. S., et al. (2013). Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication. PLoS Biol. 11:e1001604. doi: 10.1371/journal.pbio.1001604

PubMed Abstract | Crossref Full Text | Google Scholar

Gao, X., Gao, L. F., Kong, X. Q., Zhang, Y. N., Jia, S., and Meng, C. Y. (2023a). Mesenchymal stem cell-derived extracellular vesicles carrying miR-99b-3p restrain microglial activation and neuropathic pain by stimulating autophagy. Int. Immunopharmacol. 115:109695. doi: 10.1016/j.intimp.2023.109695

PubMed Abstract | Crossref Full Text | Google Scholar

Gao, X., Gao, L. F., Zhang, Y. N., Kong, X. Q., Jia, S., and Meng, C. Y. (2023b). Huc-MSCs-derived exosomes attenuate neuropathic pain by inhibiting activation of the TLR2/MyD88/NF-κB signaling pathway in the spinal microglia by targeting Rsad2. Int. Immunopharmacol. 114:109505. doi: 10.1016/j.intimp.2022.109505

PubMed Abstract | Crossref Full Text | Google Scholar

Gao, C., Yang, T., Shu, J., Gao, X., and Meng, C. (2024). Overexpression of miR-133a-3p reduces microglia activation by binding to GCH1, alleviating neuroinflammation and neuropathic pain. Exp. Brain Res. 243:23. doi: 10.1007/s00221-024-06956-y

PubMed Abstract | Crossref Full Text | Google Scholar

Gaudet, A. D., Mandrekar-Colucci, S., Hall, J. C., Sweet, D. R., Schmitt, P. J., Xu, X., et al. (2016). MiR-155 deletion in mice overcomes neuron-intrinsic and neuron-extrinsic barriers to spinal cord repair. J. Neurosci. 36, 8516–8532. doi: 10.1523/JNEUROSCI.0735-16.2016

PubMed Abstract | Crossref Full Text | Google Scholar

Ghosh, S., Dey, A., Chakrabarti, A., Bhuniya, T., Indu, N., Hait, A., et al. (2024). The theragnostic advances of exosomes in managing leukaemia. J. Cell. Mol. Med. 28:e70052. doi: 10.1111/jcmm.70052

PubMed Abstract | Crossref Full Text | Google Scholar

Golmakani, H., Azimian, A., and Golmakani, E. (2024). Newly discovered functions of miRNAs in neuropathic pain: transitioning from recent discoveries to innovative underlying mechanisms. Mol. Pain 20:17448069231225845. doi: 10.1177/17448069231225845

PubMed Abstract | Crossref Full Text | Google Scholar

Gong, Z., Zhou, D., Wu, D., Han, Y., Yu, H., Shen, H., et al. (2025). Challenges and material innovations in drug delivery to central nervous system tumors. Biomaterials 319:123180. doi: 10.1016/j.biomaterials.2025.123180

PubMed Abstract | Crossref Full Text | Google Scholar

Gotoh, S., Kawabori, M., Yamaguchi, S., Nakahara, Y., Yoshie, E., Konno, K., et al. (2025). Intranasal administration of stem cell-derived exosome alleviates cognitive impairment against subarachnoid hemorrhage. Exp. Neurol. 386:115143. doi: 10.1016/j.expneurol.2025.115143

PubMed Abstract | Crossref Full Text | Google Scholar

Graner, M. W. (2019). Roles of extracellular vesicles in high-grade gliomas: tiny particles with outsized influence. Annu. Rev. Genomics Hum. Genet. 20, 331–357. doi: 10.1146/annurev-genom-083118-015324

PubMed Abstract | Crossref Full Text | Google Scholar

Guo, Y., Zeng, J., Zhuang, Y., Jiang, C., and Xie, W. (2024). MiR-503-5p alleviates peripheral neuropathy-induced neuropathic pain in T2DM mice by regulating SEPT9 to inhibit astrocyte activation. Sci. Rep. 14:14361. doi: 10.1038/s41598-024-65096-z

PubMed Abstract | Crossref Full Text | Google Scholar

Habib, A., Liang, Y., and Zhu, N. (2023). Exosomes multifunctional roles in HIV-1: insight into the immune regulation, vaccine development and current progress in delivery system. Front. Immunol. 14:1249133. doi: 10.3389/fimmu.2023.1249133

PubMed Abstract | Crossref Full Text | Google Scholar

Han, Q., and Xu, X. M. (2020). Neurotrophin-3-mediated locomotor recovery: a novel therapeutic strategy targeting lumbar neural circuitry after spinal cord injury. Neural Regen. Res. 15, 2241–2242. doi: 10.4103/1673-5374.284985

PubMed Abstract | Crossref Full Text | Google Scholar

Haroon, K., Zheng, H., Wu, S., Liu, Z., Tang, Y., Yang, G. Y., et al. (2024). Engineered exosomes mediated targeted delivery of neuroprotective peptide NR2B9c for the treatment of traumatic brain injury. Int. J. Pharm. 649:123656. doi: 10.1016/j.ijpharm.2023.123656

PubMed Abstract | Crossref Full Text | Google Scholar

Harrell, C. R., Volarevic, V., Djonov, V., and Volarevic, A. (2022). Therapeutic potential of exosomes derived from adipose tissue-sourced mesenchymal stem cells in the treatment of neural and retinal diseases. Int. J. Mol. Sci. 23:4487. doi: 10.3390/ijms23094487

PubMed Abstract | Crossref Full Text | Google Scholar

Hassanzadeh, A., Rahman, H. S., Markov, A., Endjun, J. J., Zekiy, A. O., Chartrand, M. S., et al. (2021). Mesenchymal stem/stromal cell-derived exosomes in regenerative medicine and cancer; overview of development, challenges, and opportunities. Stem Cell Res Ther 12:297. doi: 10.1186/s13287-021-02378-7

PubMed Abstract | Crossref Full Text | Google Scholar

Horbay, R., Hamraghani, A., Ermini, L., Holcik, S., Beug, S. T., and Yeganeh, B. (2022). Role of ceramides and lysosomes in extracellular vesicle biogenesis, cargo sorting and release. Int. J. Mol. Sci. 23:15317. doi: 10.3390/ijms232315317

PubMed Abstract | Crossref Full Text | Google Scholar

Hou, J., Deng, Q., Deng, X., Zhong, W., Liu, S., and Zhong, Z. (2021). MicroRNA-146a-5p alleviates lipopolysaccharide-induced NLRP3 inflammasome injury and pro-inflammatory cytokine production via the regulation of TRAF6 and IRAK1 in human umbilical vein endothelial cells (HUVECs). Ann. Transl. Med. 9:1433. doi: 10.21037/atm-21-3903

PubMed Abstract | Crossref Full Text | Google Scholar

Hu, Y. W., Jiang, J. J., Yan, G., Wang, R. Y., and Tu, G. J. (2016). MicroRNA-210 promotes sensory axon regeneration of adult mice in vivo and in vitro. Neurosci. Lett. 622, 61–66. doi: 10.1016/j.neulet.2016.04.034

Crossref Full Text | Google Scholar

Hua, T., Yang, M., Song, H., Kong, E., Deng, M., Li, Y., et al. (2022). Huc-MSCs-derived exosomes attenuate inflammatory pain by regulating microglia pyroptosis and autophagy via the miR-146a-5p/TRAF6 axis. J. Nanobiotechnology 20:324. doi: 10.1186/s12951-022-01522-6

PubMed Abstract | Crossref Full Text | Google Scholar

Huang, J. H., Xu, Y., Yin, X. M., and Lin, F. Y. (2020). Exosomes derived from miR-126-modified MSCs promote angiogenesis and neurogenesis and attenuate apoptosis after spinal cord injury in rats. Neuroscience 424, 133–145. doi: 10.1016/j.neuroscience.2019.10.043

PubMed Abstract | Crossref Full Text | Google Scholar

Huh, Y., Ji, R. R., and Chen, G. (2017). Neuroinflammation, bone marrow stem cells, and chronic pain. Front. Immunol. 8:1014. doi: 10.3389/fimmu.2017.01014

PubMed Abstract | Crossref Full Text | Google Scholar

Hushmandi, K., Saadat, S. H., Raei, M., Aref, A. R., Reiter, R. J., Nabavi, N., et al. (2024). The science of exosomes: understanding their formation, capture, and role in cellular communication. Pathol. Res. Pract. 259:155388. doi: 10.1016/j.prp.2024.155388

PubMed Abstract | Crossref Full Text | Google Scholar

Ishii, N., and Tateno, H. (2025). Preparation of small extracellular vesicles using sequential ultrafiltration with regenerated cellulose membranes of different molecular weight cutoffs: a study of morphology and size by electron microscopy. Microsc. Microanal. 31:ozae133. doi: 10.1093/mam/ozae133

PubMed Abstract | Crossref Full Text | Google Scholar

Jia, S., Chen, G., Liang, Y., Liang, X., and Meng, C. (2021). GCH1-regulated miRNAs are potential targets for microglial activation in neuropathic pain. Biosci. Rep. 41:BSR20210051. doi: 10.1042/BSR20210051

PubMed Abstract | Crossref Full Text | Google Scholar

Jiang, Z., and Zhang, J. (2021). Mesenchymal stem cell-derived exosomes containing miR-145-5p reduce inflammation in spinal cord injury by regulating the TLR4/NF-κB signaling pathway. Cell Cycle 20, 993–1009. doi: 10.1080/15384101.2021.1919825

PubMed Abstract | Crossref Full Text | Google Scholar

Ju, Y., Bai, H., Ren, L., and Zhang, L. (2021). The role of exosome and the ESCRT pathway on enveloped virus infection. Int. J. Mol. Sci. 22:9060. doi: 10.3390/ijms22169060

PubMed Abstract | Crossref Full Text | Google Scholar

Kang, J., and Guo, Y. (2022). Human umbilical cord mesenchymal stem cells derived exosomes promote neurological function recovery in a rat spinal cord injury model. Neurochem. Res. 47, 1532–1540. doi: 10.1007/s11064-022-03545-9

PubMed Abstract | Crossref Full Text | Google Scholar

Kar, A. N., Lee, S. J., Sahoo, P. K., Thames, E., Yoo, S., Houle, J. D., et al. (2021). MicroRNAs 21 and 199a-3p regulate axon growth potential through modulation of Pten and mTor mRNAs. eNeuro. 8:ENEURO.0155-21.2021. doi: 10.1523/ENEURO.0155-21.2021

PubMed Abstract | Crossref Full Text | Google Scholar

Karri, J., Doan, J., Vangeison, C., Catalanotto, M., Nagpal, A. S., and Li, S. (2022). Emerging evidence for intrathecal management of neuropathic pain following spinal cord injury. Front. Pain Res. 3:933422. doi: 10.3389/fpain.2022.933422

PubMed Abstract | Crossref Full Text | Google Scholar

Kaye, A. D., Perilloux, D. M., Hawkins, A. M., Wester, G. C., Ragaland, A. R., Hebert, S. V., et al. (2024). Tumor necrosis factor and interleukin modulators for pathologic pain states: a narrative review. Pain Ther. 13, 481–493. doi: 10.1007/s40122-024-00603-8

PubMed Abstract | Crossref Full Text | Google Scholar

Keefe, K. M., Sheikh, I. S., and Smith, G. M. (2017). Targeting neurotrophins to specific populations of neurons: NGF, BDNF, and NT-3 and their relevance for treatment of spinal cord injury. Int. J. Mol. Sci. 18:548. doi: 10.3390/ijms18030548

PubMed Abstract | Crossref Full Text | Google Scholar

Khan, S. U., Khan, M. I., Khan, M. U., Khan, N. M., Bungau, S., and Hassan, S. S. U. (2022). Applications of extracellular vesicles in nervous system disorders: an overview of recent advances. Bioengineering 10:51. doi: 10.3390/bioengineering10010051

Crossref Full Text | Google Scholar

Kim, D. M., and Nimigean, C. M. (2016). Voltage-gated potassium channels: a structural examination of selectivity and gating. Cold Spring Harb. Perspect. Biol. 8:a029231. doi: 10.1101/cshperspect.a029231

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, H. I., Park, J., Zhu, Y., Wang, X., Han, Y., and Zhang, D. (2024). Recent advances in extracellular vesicles for therapeutic cargo delivery. Exp. Mol. Med. 56, 836–849. doi: 10.1038/s12276-024-01201-6

PubMed Abstract | Crossref Full Text | Google Scholar

Larios, J., Mercier, V., Roux, A., and Gruenberg, J. (2020). ALIX-and ESCRT-III-dependent sorting of tetraspanins to exosomes. J. Cell Biol. 219:e201904113. doi: 10.1083/jcb.201904113

Crossref Full Text | Google Scholar

Leão Nunes Filho, M. J., Barreto, E. S. R., Antunes Júnior, C. R., Alencar, V. B., Falcão Lins-Kusterer, L. E., Azi, L., et al. (2024). Efficacy of antidepressants in the treatment of chronic nonspecific low back pain: a systematic review and meta-analysis. Pain Manag. 14, 437–451. doi: 10.1080/17581869.2024.2408215

PubMed Abstract | Crossref Full Text | Google Scholar

Leinders, M., Üçeyler, N., Thomann, A., and Sommer, C. (2017). Aberrant microRNA expression in patients with painful peripheral neuropathies. J. Neurol. Sci. 380, 242–249. doi: 10.1016/j.jns.2017.07.041

PubMed Abstract | Crossref Full Text | Google Scholar

Lerussi, G., Villagrasa-Araya, V., Moltó-Abad, M., Del Toro, M., Pintos-Morell, G., Seras-Franzoso, J., et al. (2025). Extracellular vesicles as tools for crossing the blood-brain barrier to treat lysosomal storage diseases. Life 15:70. doi: 10.3390/life15010070

PubMed Abstract | Crossref Full Text | Google Scholar

Leung, A., Shirvalkar, P., Chen, R., Kuluva, J., Vaninetti, M., Bermudes, R., et al. (2020). Transcranial magnetic stimulation for pain, headache, and comorbid depression: INS-NANS expert consensus panel review and recommendation. Neuromodulation 23, 267–290. doi: 10.1111/ner.13094

PubMed Abstract | Crossref Full Text | Google Scholar

Levy, D., Jeyaram, A., Born, L. J., Chang, K. H., Abadchi, S. N., Hsu, A. T. W., et al. (2023). Impact of storage conditions and duration on function of native and cargo-loaded mesenchymal stromal cell extracellular vesicles. Cytotherapy 25, 502–509. doi: 10.1016/j.jcyt.2022.11.006

PubMed Abstract | Crossref Full Text | Google Scholar

Li, H. J., Pan, Y. B., Sun, Z. L., Sun, Y. Y., Yang, X. T., and Feng, D. F. (2018). Inhibition of miR-21 ameliorates excessive astrocyte activation and promotes axon regeneration following optic nerve crush. Neuropharmacology 137, 33–49. doi: 10.1016/j.neuropharm.2018.04.028

PubMed Abstract | Crossref Full Text | Google Scholar

Li, S., Tian, T., Zhang, T., Lin, Y., and Cai, X. (2025). A bioswitchable delivery system for microRNA therapeutics based on a tetrahedral DNA nanostructure. Nat. Protoc. 20, 336–362. doi: 10.1038/s41596-024-01050-7

PubMed Abstract | Crossref Full Text | Google Scholar

Li, H., Wan, H. Q., Zhao, H. J., Luan, S. X., and Zhang, C. G. (2019). Identification of candidate genes and miRNAs associated with neuropathic pain induced by spared nerve injury. Int. J. Mol. Med. 44, 1205–1218. doi: 10.3892/ijmm.2019.4305

PubMed Abstract | Crossref Full Text | Google Scholar

Li, X., Wei, Z., and Xue, C. (2022). Oral cell-targeted delivery systems constructed of edible materials: advantages and challenges. Molecules 27:7991. doi: 10.3390/molecules27227991

PubMed Abstract | Crossref Full Text | Google Scholar

Li, Z., Zhou, Y., and Li, Z. (2022). NFKB1 signalling activation contributes to TRPV1 over-expression via repressing MiR-375 and MiR-455: a study on neuropathic low Back pain. Folia Biol. 68, 105–111. doi: 10.14712/fb2022068030105

PubMed Abstract | Crossref Full Text | Google Scholar

Li, J., Zhu, Y., Ma, Z., Liu, Y., Sun, Z., and Wu, Y. (2021). miR-140 ameliorates neuropathic pain in CCI rats by targeting S1PR1. J. Recept. Signal Transduct. Res. 41, 401–407. doi: 10.1080/10799893.2020.1818091

PubMed Abstract | Crossref Full Text | Google Scholar

Liang, Y., Wu, J. H., Zhu, J. H., and Yang, H. (2022). Exosomes secreted by hypoxia-pre-conditioned adipose-derived mesenchymal stem cells reduce neuronal apoptosis in rats with spinal cord injury. J. Neurotrauma 39, 701–714. doi: 10.1089/neu.2021.0290

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, X., Cao, Y., Wang, S., Liu, J., and Hao, H. (2024). Extracellular vesicles: powerful candidates in nano-drug delivery systems. Drug Deliv. Transl. Res. 14, 295–311. doi: 10.1007/s13346-023-01411-x

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, Z. Y., Song, Z. W., Guo, S. W., He, J. S., Wang, S. Y., Zhu, J. G., et al. (2019). CXCL12/CXCR4 signaling contributes to neuropathic pain via central sensitization mechanisms in a rat spinal nerve ligation model. CNS Neurosci. Ther. 25, 922–936. doi: 10.1111/cns.13128

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, M., Wang, Y., Zhang, Y., Hu, D., Tang, L., Zhou, B., et al. (2025). Landscape of small nucleic acid therapeutics: moving from the bench to the clinic as next-generation medicines. Signal Transduct. Target. Ther. 10:73. doi: 10.1038/s41392-024-02112-8

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, X., Zhang, L., Xu, Z., Xiong, X., Yu, Y., Wu, H., et al. (2022). A functionalized collagen-I scaffold delivers microRNA 21-loaded exosomes for spinal cord injury repair. Acta Biomater. 154, 385–400. doi: 10.1016/j.actbio.2022.10.027

PubMed Abstract | Crossref Full Text | Google Scholar

Logan, C. J., Staton, C. C., Oliver, J. T., Bouffard, J., Kazmirchuk, T. D. D., Magi, M., et al. (2024). Thermotolerance in S. cerevisiae as a model to study extracellular vesicle biology. J Extracell Vesicles. 13:e12431. doi: 10.1002/jev2.12431

PubMed Abstract | Crossref Full Text | Google Scholar

Lotfy, A., AboQuella, N. M., and Wang, H. (2023). Mesenchymal stromal/stem cell (MSC)-derived exosomes in clinical trials. Stem Cell Res Ther 14:66. doi: 10.1186/s13287-023-03287-7

PubMed Abstract | Crossref Full Text | Google Scholar

Lv, F., Huang, Y., Lv, W., Yang, L., Li, F., Fan, J., et al. (2017). MicroRNA-146a ameliorates inflammation via TRAF6/NF-κB pathway in intervertebral disc cells. Med. Sci. Monit. 23, 659–664. doi: 10.12659/MSM.898660

PubMed Abstract | Crossref Full Text | Google Scholar

Manville, R. W., Papanikolaou, M., and Abbott, G. W. (2018). Direct neurotransmitter activation of voltage-gated potassium channels. Nat. Commun. 9:1847. doi: 10.1038/s41467-018-04266-w

PubMed Abstract | Crossref Full Text | Google Scholar

Marjani, A. A., Nader, N. D., and Aghanejad, A. (2024). Exosomes as targeted diagnostic biomarkers: recent studies and trends. Life Sci. 354:122985. doi: 10.1016/j.lfs.2024.122985

PubMed Abstract | Crossref Full Text | Google Scholar

Marofi, F., Vahedi, G., Biglari, A., Esmaeilzadeh, A., and Athari, S. S. (2017). Mesenchymal stromal/stem cells: a new era in the cell-based targeted gene therapy of cancer. Front. Immunol. 8:1770. doi: 10.3389/fimmu.2017.01770

PubMed Abstract | Crossref Full Text | Google Scholar

Mead, B., and Tomarev, S. (2017). Bone marrow-derived mesenchymal stem cells-derived exosomes promote survival of retinal ganglion cells through miRNA-dependent mechanisms. Stem Cells Transl. Med. 6, 1273–1285. doi: 10.1002/sctm.16-0428

PubMed Abstract | Crossref Full Text | Google Scholar

Meldolesi, J. (2018). Exosomes and ectosomes in intercellular communication. Curr. Biol. 28, R435–R444. doi: 10.1016/j.cub.2018.01.059

PubMed Abstract | Crossref Full Text | Google Scholar

Moisset, X., Pagé, M. G., Pereira, B., and Choinière, M. (2022). Pharmacological treatments of neuropathic pain: real-life comparisons using propensity score matching. Pain 163, 964–974. doi: 10.1097/j.pain.0000000000002461

PubMed Abstract | Crossref Full Text | Google Scholar

Mou, C., Li, Z., Liu, N., Ni, L., and Xu, Y. (2023). Low level TGF-β1-treated umbilical mesenchymal stem cells attenuates microgliosis and neuropathic pain in chronic constriction injury by exosomes/lncRNA UCA1/miR-96-5p/FOXO3a. Biochem. Biophys. Rep. 34:101477. doi: 10.1016/j.bbrep.2023.101477

PubMed Abstract | Crossref Full Text | Google Scholar

Mukherjee, A., Verma, A., Das, T., Ghosh, B., and Ghosh, Z. (2025). Circulating microRNAs in body fluid: “fingerprint” RNA snippets deeply impact reproductive biology. Reprod. Sci. 32, 555–574. doi: 10.1007/s43032-024-01753-y

PubMed Abstract | Crossref Full Text | Google Scholar

Nie, H., and Jiang, Z. (2021). Bone mesenchymal stem cell-derived extracellular vesicles deliver microRNA-23b to alleviate spinal cord injury by targeting toll-like receptor TLR4 and inhibiting NF-κB pathway activation. Bioengineered 12, 8157–8172. doi: 10.1080/21655979.2021.1977562

PubMed Abstract | Crossref Full Text | Google Scholar

Ning, X. J., Lu, X. H., Luo, J. C., Chen, C., Gao, Q., Li, Z. Y., et al. (2020). Molecular mechanism of microRNA-21 promoting Schwann cell proliferation and axon regeneration during injured nerve repair. RNA Biol. 17, 1508–1519. doi: 10.1080/15476286.2020.1777767

PubMed Abstract | Crossref Full Text | Google Scholar

Nouri, Z., Barfar, A., Perseh, S., Motasadizadeh, H., Maghsoudian, S., Fatahi, Y., et al. (2024). Exosomes as therapeutic and drug delivery vehicle for neurodegenerative diseases. J Nanobiotechnology. 22:463. doi: 10.1186/s12951-024-02681-4

PubMed Abstract | Crossref Full Text | Google Scholar

Palakurthi, S. S., Shah, B., Kapre, S., Charbe, N., Immanuel, S., Pasham, S., et al. (2024). A comprehensive review of challenges and advances in exosome-based drug delivery systems. Nanoscale Adv. 6, 5803–5826. doi: 10.1039/D4NA00501E

PubMed Abstract | Crossref Full Text | Google Scholar

Pan, Z., Shan, Q., Gu, P., Wang, X. M., Tai, L. W., Sun, M., et al. (2018). miRNA-23a/CXCR4 regulates neuropathic pain via directly targeting TXNIP/NLRP3 inflammasome axis. J. Neuroinflammation 15:29. doi: 10.1186/s12974-018-1073-0

PubMed Abstract | Crossref Full Text | Google Scholar

Parada, N., Romero-Trujillo, A., Georges, N., and Alcayaga-Miranda, F. (2021). Camouflage strategies for therapeutic exosomes evasion from phagocytosis. J. Adv. Res. 31, 61–74. doi: 10.1016/j.jare.2021.01.001

PubMed Abstract | Crossref Full Text | Google Scholar

Patil, S. M., Sawant, S. S., and Kunda, N. K. (2020). Exosomes as drug delivery systems: a brief overview and progress update. Eur. J. Pharm. Biopharm. 154, 259–269. doi: 10.1016/j.ejpb.2020.07.026

PubMed Abstract | Crossref Full Text | Google Scholar

Pedder, J. H., Sonabend, A. M., Cearns, M. D., Michael, B. D., Zakaria, R., Heimberger, A. B., et al. (2025). Crossing the blood-brain barrier: emerging therapeutic strategies for neurological disease. Lancet Neurol. 24, 246–260. doi: 10.1016/S1474-4422(24)00476-9

PubMed Abstract | Crossref Full Text | Google Scholar

Peng, C., Li, L., Zhang, M. D., Bengtsson Gonzales, C., Parisien, M., Belfer, I., et al. (2017). MiR-183 cluster scales mechanical pain sensitivity by regulating basal and neuropathic pain genes. Science 356, 1168–1171. doi: 10.1126/science.aam7671

PubMed Abstract | Crossref Full Text | Google Scholar

Picchio, V., Pontecorvi, V., Dhori, X., Bordin, A., Floris, E., Cozzolino, C., et al. (2025). The emerging role of artificial intelligence applied to exosome analysis: from cancer biology to other biomedical fields. Life Sci. 375:123752. doi: 10.1016/j.lfs.2025.123752

PubMed Abstract | Crossref Full Text | Google Scholar

Poongodi, R., Yang, T. H., Huang, Y. H., Yang, K. D., Chen, H. Z., Chu, T. Y., et al. (2024). Stem cell exosome-loaded Gelfoam improves locomotor dysfunction and neuropathic pain in a rat model of spinal cord injury. Stem Cell Res Ther 15:143. doi: 10.1186/s13287-024-03758-5

PubMed Abstract | Crossref Full Text | Google Scholar

Qi, R., Cao, J., Sun, Y., Li, Y., Huang, Z., Jiang, D., et al. (2022). Histone methylation-mediated microRNA-32-5p down-regulation in sensory neurons regulates pain behaviors via targeting Cav3.2 channels. Proc. Natl. Acad. Sci. U.S.A. 119:e2117209119. doi: 10.1073/pnas.2117209119

PubMed Abstract | Crossref Full Text | Google Scholar

Qiao, L., Hu, J., Qiu, X., Wang, C., Peng, J., Zhang, C., et al. (2023). LAMP2A, LAMP2B and LAMP2C: similar structures, divergent roles. Autophagy 19, 2837–2852. doi: 10.1080/15548627.2023.2235196

PubMed Abstract | Crossref Full Text | Google Scholar

Ren, X., Xu, R., Xu, C., and Su, J. (2024). Harnessing exosomes for targeted therapy: strategy and application. Biomater. Transl. 5, 46–58. doi: 10.12336/biomatertransl.2024.01.005

PubMed Abstract | Crossref Full Text | Google Scholar

Røikjer, J., Borbjerg, M. K., Andresen, T., Giordano, R., Hviid, C. V. B., Mørch, C. D., et al. (2024). Diabetic peripheral neuropathy: emerging treatments of neuropathic pain and novel diagnostic methods. J. Diabetes Sci. Technol. :19322968241279553. doi: 10.1177/19322968241279553

Crossref Full Text | Google Scholar

Saeedi, S., Israel, S., Nagy, C., and Turecki, G. (2019). The emerging role of exosomes in mental disorders. Transl. Psychiatry 9:122. doi: 10.1038/s41398-019-0459-9

PubMed Abstract | Crossref Full Text | Google Scholar

Sakai, A., Saitow, F., Maruyama, M., Miyake, N., Miyake, K., Shimada, T., et al. (2017). MicroRNA cluster miR-17-92 regulates multiple functionally related voltage-gated potassium channels in chronic neuropathic pain. Nat. Commun. 8:16079. doi: 10.1038/ncomms16079

PubMed Abstract | Crossref Full Text | Google Scholar

Sakai, A., Saitow, F., Miyake, N., Miyake, K., Shimada, T., and Suzuki, H. (2013). Mir-7a alleviates the maintenance of neuropathic pain through regulation of neuronal excitability. Brain 136, 2738–2750. doi: 10.1093/brain/awt191

PubMed Abstract | Crossref Full Text | Google Scholar

Sanchez, D. N. R., Bertanha, M., Fernandes, T. D., Resende, L. A. L., Deffune, E., and Amorim, R. M. (2017). Effects of canine and murine mesenchymal stromal cell transplantation on peripheral nerve regeneration. Int. J. Stem Cells 10, 83–92. doi: 10.15283/ijsc16037

PubMed Abstract | Crossref Full Text | Google Scholar

Savelieff, M. G., Elafros, M. A., Viswanathan, V., Jensen, T. S., Bennett, D. L., and Feldman, E. L. (2025). The global and regional burden of diabetic peripheral neuropathy. Nat. Rev. Neurol. 21, 17–31. doi: 10.1038/s41582-024-01041-y

Crossref Full Text | Google Scholar

Sen, S., Xavier, J., Kumar, N., Ahmad, M. Z., and Ranjan, O. P. (2023). Exosomes as natural nanocarrier-based drug delivery system: recent insights and future perspectives. 3 Biotech 13:101. doi: 10.1007/s13205-023-03521-2

PubMed Abstract | Crossref Full Text | Google Scholar

Shaman, J. A. (2024). The future of pharmacogenomics: integrating epigenetics, nutrigenomics, and beyond. J. Pers. Med. 14:1121. doi: 10.3390/jpm14121121

PubMed Abstract | Crossref Full Text | Google Scholar

Shen, W. S., Li, C. F., Zhou, Z. S., Zhai, N. N., and Pan, L. P. (2020). MicroRNA-204 silencing relieves pain of cervical spondylotic radiculopathy by targeting GDNF. Gene Ther. 27, 254–265. doi: 10.1038/s41434-019-0114-3

PubMed Abstract | Crossref Full Text | Google Scholar

Shen, F., Zheng, H., Zhou, L., Li, W., Zhang, Y., and Xu, X. (2019). LINC00657 expedites neuropathic pain development by modulating miR-136/ZEB1 axis in a rat model. J. Cell. Biochem. 120, 1000–1010. doi: 10.1002/jcb.27466

PubMed Abstract | Crossref Full Text | Google Scholar

Shityakov, S., Nagai, M., Ergün, S., Braunger, B. M., and Förster, C. Y. (2022). The protective effects of neurotrophins and microRNA in diabetic retinopathy, nephropathy and heart failure via regulating endothelial function. Biomol. Ther. 12:1113. doi: 10.3390/biom12081113

PubMed Abstract | Crossref Full Text | Google Scholar

Simeoli, R., Montague, K., Jones, H. R., Castaldi, L., Chambers, D., Kelleher, J. H., et al. (2017). Exosomal cargo including microRNA regulates sensory neuron to macrophage communication after nerve trauma. Nat. Commun. 8:1778. doi: 10.1038/s41467-017-01841-5

PubMed Abstract | Crossref Full Text | Google Scholar

Sohrabi, B., Dayeri, B., Zahedi, E., Khoshbakht, S., Nezamabadi Pour, N., Ranjbar, H., et al. (2022). Mesenchymal stem cell (MSC)-derived exosomes as novel vehicles for delivery of miRNAs in cancer therapy. Cancer Gene Ther. 29, 1105–1116. doi: 10.1038/s41417-022-00427-8

PubMed Abstract | Crossref Full Text | Google Scholar

Su, S., Shao, J., Zhao, Q., Ren, X., Cai, W., Li, L., et al. (2017). MiR-30b attenuates neuropathic pain by regulating voltage-gated sodium channel Nav1.3 in rats. Front. Mol. Neurosci. 10:126. doi: 10.3389/fnmol.2017.00126

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, L., Xia, R., Jiang, J., Wen, T., Huang, Z., Qian, R., et al. (2021). MicroRNA-96 is required to prevent allodynia by repressing voltage-gated sodium channels in spinal cord. Prog. Neurobiol. 202:102024. doi: 10.1016/j.pneurobio.2021.102024

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, W., Zhang, L., and Li, R. (2017). Overexpression of miR-206 ameliorates chronic constriction injury-induced neuropathic pain in rats via the MEK/ERK pathway by targeting brain-derived neurotrophic factor. Neurosci. Lett. 646, 68–74. doi: 10.1016/j.neulet.2016.12.047

PubMed Abstract | Crossref Full Text | Google Scholar

Tang, X. H., Guo, T., Gao, X. Y., Wu, X. L., Xing, X. F., Ji, J. F., et al. (2021). Exosome-derived noncoding RNAs in gastric cancer: functions and clinical applications. Mol. Cancer 20:99. doi: 10.1186/s12943-021-01396-6

PubMed Abstract | Crossref Full Text | Google Scholar

Tavares-Ferreira, D., Lawless, N., Bird, E. V., Atkins, S., Collier, D., Sher, E., et al. (2019). Correlation of miRNA expression with intensity of neuropathic pain in man. Mol. Pain 15:1744806919860323. doi: 10.1177/1744806919860323

PubMed Abstract | Crossref Full Text | Google Scholar

Théry, C., Witwer, K. W., Aikawa, E., Alcaraz, M. J., Anderson, J. D., Andriantsitohaina, R., et al. (2018). Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell Vesicles. 7:1535750. doi: 10.1080/20013078.2018.1535750

PubMed Abstract | Crossref Full Text | Google Scholar

van Battum, E. Y., Verhagen, M. G., Vangoor, V. R., Fujita, Y., Derijck, A., O’Duibhir, E., et al. (2018). An image-based miRNA screen identifies miRNA-135s as regulators of CNS axon growth and regeneration by targeting Krüppel-like factor 4. J. Neurosci. 38, 613–630. doi: 10.1523/JNEUROSCI.0662-17.2017

Crossref Full Text | Google Scholar

Vranken, J. H. (2012). Elucidation of pathophysiology and treatment of neuropathic pain. Cent. Nerv. Syst. Agents Med. Chem. 12, 304–314. doi: 10.2174/187152412803760645

Crossref Full Text | Google Scholar

Vučemilović, A. (2024). Exosomes: intriguing mediators of intercellular communication in the organism’s response to noxious agents. Arh. Hig. Rada Toksikol. 75, 228–239. doi: 10.2478/aiht-2024-75-3923

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, H., Li, Q., Zou, J., Shu, J., Zhang, A., Zhang, H., et al. (2024). Mapping the research landscape of microRNAs in pain: a comprehensive bibliometric analysis. Front. Mol. Neurosci. 17:1493822. doi: 10.3389/fnmol.2024.1493822

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, Z., Liu, F., Wei, M., Qiu, Y., Ma, C., Shen, L., et al. (2018). Chronic constriction injury-induced microRNA-146a-5p alleviates neuropathic pain through suppression of IRAK1/TRAF6 signaling pathway. J. Neuroinflammation 15:179. doi: 10.1186/s12974-018-1215-4

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, Y., and Wu, X. (2024). New perspectives and prospects of microRNA delivery in diabetic wound healing. Mol. Pharmacol. 106, 84–91. doi: 10.1124/molpharm.124.000899

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, C., Xu, M., Fan, Q., Li, C., and Zhou, X. (2023). Therapeutic potential of exosome-based personalized delivery platform in chronic inflammatory diseases. Asian J. Pharm. Sci. 18:100772. doi: 10.1016/j.ajps.2022.100772

PubMed Abstract | Crossref Full Text | Google Scholar

Wilkerson, J. L., Jiang, J., Felix, J. S., Bray, J. K., da Silva, L., Gharaibeh, R. Z., et al. (2020). Alterations in mouse spinal cord and sciatic nerve microRNAs after the chronic constriction injury (CCI) model of neuropathic pain. Neurosci. Lett. 731:135029. doi: 10.1016/j.neulet.2020.135029

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, H., Zhu, L., Geng, X., Guo, X., Wang, T., Xu, J., et al. (2025). miR-363-5p protects from neuropathic pain in chronic constriction injury (CCI) rat models and regulates Schwann cell injury via negatively modulating SERPING1. Neurol. Res. 47, 35–43. doi: 10.1080/01616412.2024.2438613

PubMed Abstract | Crossref Full Text | Google Scholar

Xia, B., Gao, J., Li, S., Huang, L., Zhu, L., Ma, T., et al. (2020). Mechanical stimulation of Schwann cells promote peripheral nerve regeneration via extracellular vesicle-mediated transfer of microRNA 23b-3p. Theranostics 10, 8974–8995. doi: 10.7150/thno.44912

PubMed Abstract | Crossref Full Text | Google Scholar

Xin, H., Katakowski, M., Wang, F., Qian, J. Y., Liu, X. S., Ali, M. M., et al. (2017). MicroRNA cluster miR-17-92 cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats. Stroke 48, 747–753. doi: 10.1161/STROKEAHA.116.015204

PubMed Abstract | Crossref Full Text | Google Scholar

Xing, Y. H., Ren, X. S., Li, D. H., and Liu, L. (2025). Exosome separation and analysis based on microfluidics technology and its clinical applications. Se Pu 43, 455–471. doi: 10.3724/SP.J.1123.2024.10032

PubMed Abstract | Crossref Full Text | Google Scholar

Yamayoshi, A., Oyama, S., Kishimoto, Y., Konishi, R., Yamamoto, T., Kobori, A., et al. (2020). Development of antibody-oligonucleotide complexes for targeting exosomal MicroRNA. Pharmaceutics 12:545. doi: 10.3390/pharmaceutics12060545

PubMed Abstract | Crossref Full Text | Google Scholar

Yan, X. T., Ji, L. J., Wang, Z., Wu, X., Wang, Q., Sun, S., et al. (2017). MicroRNA-93 alleviates neuropathic pain through targeting signal transducer and activator of transcription 3. Int. Immunopharmacol. 46, 156–162. doi: 10.1016/j.intimp.2017.01.027

PubMed Abstract | Crossref Full Text | Google Scholar

Yan, X. T., Zhao, Y., Cheng, X. L., He, X. H., Wang, Y., Zheng, W. Z., et al. (2018). Inhibition of miR-200b/miR-429 contributes to neuropathic pain development through targeting zinc finger E box binding protein-1. J. Cell. Physiol. 233, 4815–4824. doi: 10.1002/jcp.26284

PubMed Abstract | Crossref Full Text | Google Scholar

Yanagawa, K., Kuma, A., Hamasaki, M., Kita, S., Yamamuro, T., Nishino, K., et al. (2024). The Rubicon-WIPI axis regulates exosome biogenesis during ageing. Nat. Cell Biol. 26, 1558–1570. doi: 10.1038/s41556-024-01481-0

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, K., Fu, W., Deng, M., Li, X., Wu, M., and Wang, Y. (2024). The sphingolipids change in exosomes from cancer patients and association between exosome release and sphingolipids level based on a pseudotargeted lipidomics method. Anal. Chim. Acta 1305:342527. doi: 10.1016/j.aca.2024.342527

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, X., Huang, X., Lu, W., Yan, F., Ye, Y., Wang, L., et al. (2023). Transcriptome profiling of miRNA-mRNA interactions and associated mechanisms in chemotherapy-induced neuropathic pain. Mol. Neurobiol. 60, 5672–5690. doi: 10.1007/s12035-023-03398-5

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, T., Martin, P., Fogarty, B., Brown, A., Schurman, K., Phipps, R., et al. (2015). Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm. Res. 32, 2003–2014. doi: 10.1007/s11095-014-1593-y

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, R., Wang, Q. Q., Feng, Y., Li, X. H., Li, G. X., She, F. L., et al. (2023). Over-expression of miR-3584-5p represses Nav1.8 channel aggravating neuropathic pain caused by chronic constriction injury. Mol. Neurobiol. 60, 5237–5255. doi: 10.1007/s12035-023-03394-9

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, S., Zhu, H., Jin, H., Wang, K., Song, J., Sun, N., et al. (2025). Bio-orthogonal-labeled exosomes reveals specific distribution in vivo and provides potential application in ARDS therapy. Biomaterials 319:123208. doi: 10.1016/j.biomaterials.2025.123208

PubMed Abstract | Crossref Full Text | Google Scholar

Yao, L., Zi, G., He, M., Xu, Y., Wang, L., and Peng, B. (2025). Asparagine endopeptidase regulates lysosome homeostasis via modulating endomembrane phosphoinositide composition. Cell Death Dis. 15:883. doi: 10.1038/s41419-024-07187-3

PubMed Abstract | Crossref Full Text | Google Scholar

Ye, F., Du, L., Huang, W., and Wang, S. (2022). Shared genetic regulatory networks contribute to neuropathic and inflammatory pain: multi-omics systems analysis. Biomol. Ther. 12:1454. doi: 10.3390/biom12101454

PubMed Abstract | Crossref Full Text | Google Scholar

Ye, H., Wang, F., Xu, G., Shu, F., Fan, K., and Wang, D. (2023). Advancements in engineered exosomes for wound repair: current research and future perspectives. Front. Bioeng. Biotechnol. 11:1301362. doi: 10.3389/fbioe.2023.1301362

PubMed Abstract | Crossref Full Text | Google Scholar

Yu, T., Zhao, C., Hou, S., Zhou, W., Wang, B., and Chen, Y. (2019). Exosomes secreted from miRNA-29b-modified mesenchymal stem cells repaired spinal cord injury in rats. Braz. J. Med. Biol. Res. 52:e8735. doi: 10.1590/1414-431x20198735

PubMed Abstract | Crossref Full Text | Google Scholar

Zarezadeh Mehrabadi, A., Aghamohamadi, N., Khoshmirsafa, M., Aghamajidi, A., Pilehforoshha, M., Massoumi, R., et al. (2022). The roles of interleukin-1 receptor accessory protein in certain inflammatory conditions. Immunology 166, 38–46. doi: 10.1111/imm.13462

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, H., and Chen, H. (2021). TRPA1 involved in miR-141-5p-alleviated neuropathic pain induced by oxaliplatin. Neuroreport 32, 284–290. doi: 10.1097/WNR.0000000000001589

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, X., Guo, H., Xie, A., Liao, O., Ju, F., and Zhou, Y. (2020a). MicroRNA-144 relieves chronic constriction injury-induced neuropathic pain via targeting RASA1. Biotechnol. Appl. Biochem. 67, 294–302. doi: 10.1002/bab.1854

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, D., Lee, H., Zhu, Z., Minhas, J. K., and Jin, Y. (2017). Enrichment of selective miRNAs in exosomes and delivery of exosomal miRNAs in vitro and in vivo. Am. J. Physiol. Lung Cell. Mol. Physiol. 312, L110–L1121. doi: 10.1152/ajplung.00423.2016

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, J., Li, K., Gao, L., Zhu, P., Shu, L., Cai, L., et al. (2024). Glucose metabolism disorder related to follicular fluid exosomal miR-122-5p in cumulus cells of endometriosis patients. Reproduction 168:e240028. doi: 10.1530/REP-24-0028

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, K., Li, P., Jia, Y., Liu, M., and Jiang, J. (2023). Concise review: current understanding of extracellular vesicles to treat neuropathic pain. Front. Aging Neurosci. 15:1131536. doi: 10.3389/fnagi.2023.1131536

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y., Liu, H. L., An, L. J., Li, L., Wei, M., Ge, D. J., et al. (2019). miR-124-3p attenuates neuropathic pain induced by chronic sciatic nerve injury in rats via targeting EZH2. J. Cell. Biochem. 120, 5747–5755. doi: 10.1002/jcb.27861

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, J., Rong, L., Shao, J., Zhang, Y., Liu, Y., Zhao, S., et al. (2021). Epigenetic restoration of voltage-gated potassium channel Kv1.2 alleviates nerve injury-induced neuropathic pain. J. Neurochem. 156, 367–378. doi: 10.1111/jnc.15117

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Y. U., Ye, G., Zhao, J., Chen, Y., Kong, L., Sheng, C., et al. (2022). Exosomes carried miR-181c-5p alleviates neuropathic pain in CCI rat models. An. Acad. Bras. Cienc. 94:e20210564. doi: 10.1590/0001-3765202220210564

Crossref Full Text | Google Scholar

Zhang, X., Zhang, Y., Cai, W., Liu, Y., Liu, H., Zhang, Z., et al. (2020b). MicroRNA-128-3p alleviates neuropathic pain through targeting ZEB1. Neurosci. Lett. 729:134946. doi: 10.1016/j.neulet.2020.134946

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, F., Zhang, L., and Yu, H. (2024). Potential druggability of mesenchymal stem/stromal cell-derived exosomes. Curr. Stem Cell Res. Ther. 19, 1195–1209. doi: 10.2174/011574888X311270240319084835

PubMed Abstract | Crossref Full Text | Google Scholar

Zhao, J., Ding, Y., He, R., Huang, K., Liu, L., Jiang, C., et al. (2020). Dose-effect relationship and molecular mechanism by which BMSC-derived exosomes promote peripheral nerve regeneration after crush injury. Stem Cell Res Ther 11:360. doi: 10.1186/s13287-020-01872-8

PubMed Abstract | Crossref Full Text | Google Scholar

Zhao, M., Li, Q., Chai, Y., Rong, R., He, L., Zhang, Y., et al. (2025). An anti-CD19-exosome delivery system navigates the blood-brain barrier for targeting of central nervous system lymphoma. J. Nanobiotechnology. 23:173. doi: 10.1186/s12951-025-03238-9

PubMed Abstract | Crossref Full Text | Google Scholar

Zhong, L., Xiao, W., Wang, F., Liu, J., and Zhi, L. J. (2019). miR-21-5p inhibits neuropathic pain development via directly targeting C-C motif ligand 1 and tissue inhibitor of metalloproteinase-3. J. Cell. Biochem. 120, 16614–16623. doi: 10.1002/jcb.28920

PubMed Abstract | Crossref Full Text | Google Scholar

Zhou, X., Deng, X., Liu, M., He, M., Long, W., Xu, Z., et al. (2023). Intranasal delivery of BDNF-loaded small extracellular vesicles for cerebral ischemia therapy. J. Control. Release 357, 1–19. doi: 10.1016/j.jconrel.2023.03.033

PubMed Abstract | Crossref Full Text | Google Scholar

Glossary

Key terminology -

miRNA - microRNA

NP - Neuropathic pain

Cells & exosomes -

ADSC-EXs - Adipose-derived mesenchymal stem cell exosomes

BMSC-EVs - Bone marrow mesenchymal stem cell-derived extracellular vesicles

DRG - Dorsal root ganglion

EVs - Extracellular vesicles

huc-MSCs - Human umbilical cord mesenchymal stem cells

MSC - Mesenchymal stem cell

SKP-SCs - Skin-derived precursor Schwann cells

Clinical terms -

AEs - Adverse events

ASIA - American Spinal Injury Association

BBB - Blood-brain barrier

CNS - Central nervous system

GMP - Good manufacturing practice

MCID - Minimal clinically important difference

Disease models -

CCI - Chronic constriction injury

CIP - Chemotherapy-induced peripheral neuropathy

DPN - Diabetic peripheral neuropathy

ONI - Optic nerve injury

OXA - Oxaliplatin

pSNL - Partial sciatic nerve ligation

SCI - Spinal cord injury

SNI - Spared nerve injury

SNL - Spinal nerve ligation

TN - Trigeminal neuralgia

Molecular pathways -

Akt - Protein kinase B

BDNF - Brain-derived neurotrophic factor

EFNA3 - Ephrin-A3

GDNF - Glial cell-derived neurotrophic factor

GAP-43 - Growth-associated protein 43

IRAK1 - Interleukin-1 receptor-associated kinase 1

JNK3 - c-Jun N-terminal kinase 3

KLF4 - Krüppel-like factor 4

LAMP2B - Lysosome-associated membrane protein 2B

MAPK - Mitogen-activated protein kinase

mTOR - Mechanistic target of rapamycin

NF-κB - Nuclear factor kappa-light-chain-enhancer of activated B cells

NGF - Nerve growth factor

NLRP3 - NLR Family pyrin domain containing 3

NMDA - N-Methyl-D-aspartate receptor

NT-3 - Neurotrophin-3

PI3K - Phosphoinositide 3-kinase

PTEN - Phosphatase and tensin homolog

Rab27a/b - Ras-related protein Rab-27A/B

SEPT9 - Septin 9

STAT3 - Signal transducer and activator of transcription 3

TGF-β1 - Transforming growth factor beta 1

TLR4 - Toll-like receptor 4

TRAF6 - TNF receptor-associated factor 6

TRPA1 - Transient receptor potential ankyrin 1

TRPV1 - Transient receptor potential vanilloid 1

TXNIP - Thioredoxin-interacting protein

ZEB1 - Zinc Finger E-box binding homeobox 1

Other key terminology -

DAMPs - Damage-associated molecular patterns

lncRNA - Long non-coding rna

TDN - Tetrahedral DNA nanostructure

UCA1 - Urothelial cancer-associated 1

Technical methods -

ESCRT - Endosomal sorting complexes required for transport

GWAS - Genome-wide association study

ISEV - International Society for Extracellular Vesicles

MISEV - Minimal information for EV studies

NGS - Next-generation sequencing

SCS - Spinal cord stimulation

TENS - Transcutaneous electrical nerve stimulation

Keywords: neuropathic pain, targeted delivery, miRNA therapeutics, exosomes, pain management, clinical translation

Citation: Wei Z, Guo C, Zhou H, Wu Y, Zhou X, Chen J and Li F (2025) Exosome-mediated miRNA delivery: a molecular switch for reshaping neuropathic pain therapy. Front. Mol. Neurosci. 18:1625943. doi: 10.3389/fnmol.2025.1625943

Received: 09 May 2025; Accepted: 19 June 2025;
Published: 04 July 2025.

Edited by:

Lei Yu, Rutgers, The State University of New Jersey, United States

Reviewed by:

Parisa Gazerani, Oslo Metropolitan University, Norway
Paramita Basu, University of Pittsburgh, United States

Copyright © 2025 Wei, Guo, Zhou, Wu, Zhou, Chen and Li. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jibing Chen, amliaW5nY2hlbjM5OEAxNjMuY29t; Fujun Li, bGZqeXlxQDE2My5jb20=

ORCID: Ziqing Wei, orcid.org/0009-0000-7931-3226
Chunhui Guo, orcid.org/0009-0000-3751-6420
Hang Zhou,orcid.org/0009-0006-7067-5435
Yanling Wu, orcid.org/0009-0001-6302-8487
Jibing Chen, orcid.org/0000-0001-8596-2126
Fujun Li, orcid.org/0009-0001-9837-3678

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.